US20040053229A1 - Mammalian protein phosphatases - Google Patents

Mammalian protein phosphatases Download PDF

Info

Publication number
US20040053229A1
US20040053229A1 US10/168,506 US16850602A US2004053229A1 US 20040053229 A1 US20040053229 A1 US 20040053229A1 US 16850602 A US16850602 A US 16850602A US 2004053229 A1 US2004053229 A1 US 2004053229A1
Authority
US
United States
Prior art keywords
seq
phosphatase
polypeptide
nucleic acid
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/168,506
Inventor
Gregory Plowman
Ricardo Martinez
David Whyte
Gerard Manning
Sucha Sudarsanam
Ronald Hill
Peter Flanagan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sugen LLC
Original Assignee
Sugen LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sugen LLC filed Critical Sugen LLC
Priority to US10/168,506 priority Critical patent/US20040053229A1/en
Priority claimed from PCT/US2000/034736 external-priority patent/WO2001046394A2/en
Assigned to SUGEN, INC. reassignment SUGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PLOWMAN, GREGORY D., WHYTE, DAVID, FLANAGAN, PETER, HILL, RONALD, MANNING, GERARD, MARTINEZ, RICARDO, SUDARSANAM, SUCHA
Publication of US20040053229A1 publication Critical patent/US20040053229A1/en
Priority to US10/838,181 priority patent/US20050084877A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/042Disorders of carbohydrate metabolism, e.g. diabetes, glucose metabolism

Definitions

  • Cellular signal transduction is a fundamental mechanism whereby external stimuli that regulate diverse cellular processes are relayed to the interior of cells.
  • One of the key biochemical mechanisms of signal transduction involves the reversible phosphorylation of proteins by protein kinases, which enables regulation of the activity of mature proteins by altering their structure and function.
  • the best characterized protein kinases in eukaryotes phosphorylate proteins on the alcohol moiety of serine, threonine and tyrosine residues. These kinases largely fall into two groups: those specific for phosphorylating serines and threonines, and those specific for phosphorylating tyrosines.
  • CTP protein tyrosine phosphatases
  • DSP dual-specificity phosphatases
  • STP serine or threonine
  • Phosphatases possess a variety of non-catalytic domains that are believed to interact with upstream regulators. Examples include proline-rich domains for interaction with SH3-containing proteins, or specific domains for interaction with Rac, Rho, and Rab small G-proteins. These interactions may provide a mechanism for cross-talk between distinct biochemical pathways in response to external stimuli such as the activation of a variety of cell surface receptors, including tyrosine kinases, cytokine receptors, TNF receptor, Fas, T cell receptors, CD28, or CD40.
  • novel proteins include: eight (8) MAP kinase phosphatase enzymes (“MKPs”), which are members of the DSP family; two (2) phosphatases from the STP family; and two (2) phosphatases from the PTP family.
  • MKPs MAP kinase phosphatase enzymes
  • the classification of novel proteins as belonging to established families has proven highly accurate, not only in predicting motifs present in the remaining non-catalytic portion of each protein, but also in the regulation, substrates, and signaling pathways fo these proteins.
  • One aspect of the invention features an identified, isolated, enriched, or purified nucleic acid molecule encoding a phosphatase polypeptide, having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • isolated in reference to nucleic acid is meant a polymer of 10 (preferably 21, more preferably 39, most preferably 75) or more nucleotides conjugated to each other, including DNA and RNA that is isolated from a natural source or that is synthesized as the sense or complementary antisense strand.
  • nucleic acids are preferred, for example those of 300, 600, 900, 1200, 1500, or more nucleotides and/or those having at least 50%, 60%, 75%, 90%, 95% or 99% identity to a sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12.
  • the isolated nucleic acid of the present invention is unique in the sense that it is not found in a pure or separated state in nature.
  • Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular (i.e., chromosomal) environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment. The term does not imply that the sequence is the only nucleotide chain present, but that it is essentially free (about 90-95% pure at least) of non-nucleotide material naturally associated with it, and thus is distinguished from isolated chromosomes.
  • enriched in reference to nucleic acid is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction (2- to 5-fold) of the total DNA or RNA present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of other DNA or RNA present, or by a preferential increase in the amount of the specific DNA or RNA sequence, or by a combination of the two. However, it should be noted that enriched does not imply that there are no other DNA or RNA sequences present, just that the relative amount of the sequence of interest has been significantly increased.
  • the term “significant” is used to indicate that the level of increase is useful to the person making such an increase, and generally means an increase relative to other nucleic acids of about at least 2-fold, more preferably at least 5- to 10-fold or even more.
  • the term also does not imply that there is no DNA or RNA from other sources.
  • the DNA from other sources may, for example, comprise DNA from a yeast or bacterial genome, or a cloning vector such as pUC19. This term distinguishes from naturally occurring events, such as viral infection, or tumor-type growths, in which the level of one mRNA may be naturally increased relative to other species of mRNA. That is, the term is meant to cover only those situations in which a person has intervened to elevate the proportion of the desired nucleic acid.
  • nucleotide sequence be in purified form.
  • purified in reference to nucleic acid does not require absolute purity (such as a homogeneous preparation). Instead, it represents an indication that the sequence is relatively more pure than in the natural environment (compared to the natural level this level should be at least 2- to 5-fold greater, e.g., in terms of mg/mL).
  • Individual clones isolated from a cDNA library may be purified to electrophoretic homogeneity. The claimed DNA molecules obtained from these clones could be obtained directly from total DNA or from total RNA.
  • the cDNA clones are not naturally occurring, but rather are preferably obtained via manipulation of a partially purified naturally occurring substance (messenger RNA).
  • substitutions may include the replacement of an amino acid by a residue having similar physicochemical properties, such as substituting one aliphatic residue (Ile, Val, Leu or Ala) for another, or substitution between basic residues Lys and Arg, acidic residues Glu and Asp, amide residues Gln and Asn, hydroxyl residues Ser and Tyr, or aromatic residues Phe and Tyr.
  • amino acid exchanges which have only slight, if any, effects on the overall protein can be found in Bowie et al., Science, 1990, 247:1306-1310, which is incorporated herein by reference in its entirety including any figures, tables, or drawings. In all cases, all permutations are intended to be covered by this disclosure.
  • amino acid sequence of the phosphatase peptide of the invention will be substantially similar to a sequence having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or the corresponding full-length amino acid sequence, or fragments thereof.
  • catalytic activity defines the rate at which a phosphatase catalytic domain phosphorylates a substrate.
  • Catalytic activity can be measured, for example, by determining the amount of a substrate converted to a phosphorylated product as a function of time.
  • Catalytic activity can be measured by methods of the invention by holding time constant and determining the concentration of a phosphorylated substrate after a fixed period of time.
  • Phosphorylation of a substrate occurs at the active site of a protein phosphatase.
  • the active site is normally a cavity in which the substrate binds to the protein phosphatase and is phosphorylated.
  • the invention features isolated, enriched, or purified nucleic acid molecules encoding phosphatase polypeptides, further comprising a vector or promoter effective to initiate transcription in a host cell.
  • the invention also features recombinant nucleic acid, preferably in a cell or an organism.
  • the isolated nucleic acid comprises, consists essentially of, or consists of a nucleic acid sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, which encodes an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, a functional derivative thereof, or at least 35, 40, 45, 50, 60, 75, 100, 200, or 300 contiguous amino acids selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO:
  • unique nucleic acid region is meant a sequence present in a nucleic acid coding for a phosphatase polypeptide that is not present in a sequence coding for any other naturally occurring polypeptide.
  • Such regions preferably encode 32 (preferably 40, more preferably 45, most preferably 55) or more contiguous amino acids set forth in a full-length amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • a unique nucleic acid region is preferably of mammalian origin.
  • nucleic acid probe for the detection of nucleic acid encoding a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24 in a sample.
  • the nucleic acid probe contains a nucleotide base sequence that will hybridize to the sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or a functional derivative thereof.
  • Methods for using the probes include detecting the presence or amount of phosphatase RNA in a sample by contacting the sample with a nucleic acid probe under conditions such that hybridization occurs and detecting the presence or amount of the probe bound to phosphatase RNA.
  • the nucleic acid duplex formed between the probe and a nucleic acid sequence coding for a phosphatase polypeptide may be used in the identification of the sequence of the nucleic acid detected (Nelson et al., in Nonisotopic DNA Probe Techniques, Academic Press, San Diego, Kricka, ed., p. 275, 1992, hereby incorporated by reference herein in its entirety, including any drawings, figures, or tables).
  • Kits for performing such methods may be constructed to include a container means having disposed therein a nucleic acid probe.
  • the invention describes a recombinant cell or tissue comprising a nucleic acid molecule encoding a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the nucleic acid may be under the control of the genomic regulatory elements, or may be under the control of exogenous regulatory elements including an exogenous promoter.
  • exogenous it is meant a promoter that is not normally coupled in vivo transcriptionally to the coding sequence for the phosphatase polypeptides.
  • the polypeptide is preferably a fragment of the protein encoded by a full-length amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • fragment is meant an amino acid sequence present in a phosphatase polypeptide.
  • such a sequence comprises at least 32, 45, 50, 60, 100, 200, or 300 contiguous amino acids of a fill-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ D NO: 24.
  • a fill-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ D NO: 24.
  • the invention features an isolated, enriched, or purified phosphatase polypeptide having the amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ED NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • isolated in reference to a polypeptide is meant a polymer of 6 (preferably 12, more preferably 18, most preferably 25, 32, 40, or 50) or more amino acids conjugated to each other, including polypeptides that are isolated from a natural source or that are synthesized.
  • longer polypeptides are preferred, such as those with 100, 200, 300, 400, 450, 500, 550, 600, 700, 800, 900 or more contiguous amino acids of a full-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • isolated polypeptides of the present invention are unique in the sense that they are not found in a pure or separated state in nature.
  • Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment. The term does not imply that the sequence is the only amino acid chain present, but that it is essentially free (about 90-95% pure at least) of non-amino acid-based material naturally associated with it.
  • enriched in reference to a polypeptide is meant that the specific amino acid sequence constitutes a significantly higher fraction (2- to 5-fold) of the total amino acid sequences present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of other amino acid sequences present, or by a preferential increase in the amount of the specific amino acid sequence of interest, or by a combination of the two. However, it should be noted that enriched does not imply that there are no other amino acid sequences present, just that the relative amount of the sequence of interest has been significantly increased.
  • the phosphatase polypeptide comprises an amino acid sequence having (a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; and (b) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, except that it lacks one or more of the domains selected from the group consisting of a C-terminal catalytic domain, an N-terminal domain, a catalytic domain, a C
  • the polypeptides to be expressed in host cells may also be fusion proteins which include regions from heterologous proteins. Such regions may be included to allow, e.g., secretion, improved stability, or facilitated purification of the polypeptide.
  • a sequence encoding an appropriate signal peptide can be incorporated into expression vectors.
  • a DNA sequence for a signal peptide secretory leader
  • a signal peptide that is functional in the intended host cell promotes extracellular secretion of the polypeptide.
  • the invention features an antibody (e.g., a monoclonal or polyclonal antibody) having specific binding affinity to a phosphatase polypeptide or a phosphatase polypeptide domain or fragment where the polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • an antibody e.g., a monoclonal or polyclonal antibody having specific binding affinity to a phosphatase polypeptide or a phosphatase polypeptide domain or fragment
  • the polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO:
  • Antibodies or antibody fragments having specific binding affinity to a phosphatase polypeptide of the invention may be used in methods for detecting the presence and/or amount of phosphatase polypeptide in a sample by probing the sample with the antibody under conditions suitable for phosphatase-antibody immunocomplex formation and detecting the presence and/or amount of the antibody conjugated to the phosphatase polypeptide.
  • Diagnostic kits for performing such methods may be constructed to include antibodies or antibody fragments specific for the phosphatase as well as a conjugate of a binding partner of the antibodies or the antibodies themselves.
  • An antibody or antibody fragment with specific binding affinity to a phosphatase polypeptide of the invention can be isolated, enriched, or purified from a prokaryotic or eukaryotic organism. Routine methods known to those skilled in the art enable production of antibodies or antibody fragments, in both prokaryotic and eukaryotic organisms. Purification, enrichment, and isolation of antibodies, which are polypeptide molecules, are described above.
  • Diagnostic kits for performing such methods may be constructed to include a first container containing the antibody and a second container having a conjugate of a binding partner of the antibody and a label, such as, for example, a radioisotope.
  • the diagnostic kit may also include notification of an FDA approved use and instructions therefor.
  • negative control antibody refers to an antibody derived from similar source as the antibody having specific binding affinity, but where it displays no binding affinity to a polypeptide of the invention.
  • the invention features a phosphatase polypeptide binding agent able to bind to a phosphatase polypeptide selected from the group having (a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the binding agent is preferably a purified antibody that recognizes an epitope present on a phosphatase polypeptide of the invention.
  • Other binding agents include molecules that bind to phosphatase polypeptides and analogous molecules that bind to a phosphatase polypeptide. Such binding agents may be identified by using assays that measure phosphatase binding partner activity.
  • the invention also features a method for screening for human cells containing a phosphatase polypeptide of the invention or an equivalent sequence.
  • the method involves identifying the novel polypeptide in human cells using techniques that are routine and standard in the art, such as those described herein for identifying the phosphatases of the invention (e.g., cloning, Southern or Northern blot analysis, in situ hybridization, PCR amplification, etc.).
  • modulates refers to the ability of a compound to alter the function of a phosphatase of the invention.
  • a modulator preferably activates or inhibits the activity of a phosphatase of the invention depending on the concentration of the compound exposed to the phosphatase.
  • modulates also refers to altering the function of phosphatases of the invention by increasing or decreasing the probability that a complex forms between the phosphatase and a natural binding partner.
  • a modulator preferably increases the probability that such a complex forms between the phosphatase and the natural binding partner, more preferably increases or decreases the probability that a complex forms between the phosphatase and the natural binding partner depending on the concentration of the compound exposed to the phosphatase, and most preferably decreases the probability that a complex forms between the phosphatase and the natural binding partner.
  • complex refers to an assembly of at least two molecules bound to one another. Signal transduction complexes often contain at least two protein molecules bound to one another.
  • natural binding partner refers to polypeptides, lipids, small molecules, or nucleic acids that bind to phosphatases in cells.
  • a change in the interaction between a phosphatase and a natural binding partner can manifest itself as an increased or decreased probability that the interaction forms, or an increased or decreased concentration of phosphatase/natural binding partner complex.
  • the term “contacting” as used herein refers to mixing a solution comprising the test compound with a liquid medium bathing the cells of the methods.
  • the solution comprising the compound may also comprise another component, such as dimethyl sulfoxide (DMSO), which facilitates the uptake of the test compound or compounds into the cells of the methods.
  • DMSO dimethyl sulfoxide
  • the solution comprising the test compound may be added to the medium bathing the cells by utilizing a delivery apparatus, such as a pipette-based device or syringe-based device.
  • the invention features methods for identifying a substance that modulates phosphatase activity in a cell comprising the steps of: (a) expressing a phosphatase polypeptide in a cell, wherein said polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; (b) adding a test substance to said cell; and (c) monitoring a change in cell phenotype or the interaction between said polypeptide and a natural binding partner.
  • Another aspect of the instant invention is directed to methods of identifying compounds that bind to phosphatase polypeptides of the present invention, comprising contacting the phosphatase polypeptides with a compound, and determining whether the compound binds the phosphatase polypeptides.
  • the methods of the invention also embrace compounds that are attached to a label, such as a radiolabel (e.g., 125 I, 35 S, 32 P, 33 P, 3 H), a fluorescence label, a chemiluminescent label, an enzymic label and an immunogenic label.
  • a label such as a radiolabel (e.g., 125 I, 35 S, 32 P, 33 P, 3 H), a fluorescence label, a chemiluminescent label, an enzymic label and an immunogenic label.
  • the phosphatase polypeptides employed in such a test may either be free in solution, attached to a solid support, borne on a cell surface, located intracellularly or associated with a portion of a cell.
  • One skilled in the art can, for example, measure the formation of complexes between a phosphatase polypeptide and the compound being tested.
  • one skilled in the art can examine the diminution in complex formation between a phosphatase polypeptide
  • the present invention is particularly useful for screening compounds by using a phosphatase polypeptide in any of a variety of drug screening techniques.
  • the compounds to be screened include, but are not limited to, extracellular, intracellular, biological or chemical origin.
  • the phosphatase polypeptide employed in such a test may be in any form, preferably, free in solution, attached to a solid support, borne on a cell surface or located intracellularly.
  • One skilled in the art can, for example, measure the formation of complexes between a phosphatase polypeptide and the compound being tested.
  • one skilled in the art can examine the diminution in complex formation between a phosphatase polypeptide and its substrate caused by the compound being tested.
  • the activity of phosphatase polypeptides of the invention can be determined by, for example, examining the ability to bind or be activated by chemically synthesised peptide ligands.
  • the activity of the phosphatase polypeptides can be assayed by examining their ability to bind metal ions such as calcium, hormones, chemokines, neuropeptides, neurotransmitters, nucleotides, lipids, odorants, and photons.
  • modulators of the phosphatase polypeptide's activity may alter a phosphatase function, such as a binding property of a phosphatase or an activity such as signal transduction or membrane localization.
  • the assay may take the form of a yeast growth assay, an Aequorin assay, a Luciferase assay, a mitogenesis assay, a MAP Phosphatase activity assay, as well as other binding or function-based assays of phosphatase activity that are generally known in the art.
  • the invention includes any of the receptor and non-receptor protein tyrosine phosphatases, receptor and non-receptor protein phosphatases, polypeptides containing SRC homology 2 and 3 domains, phosphotyrosine binding proteins (SRC homology 2 (SH2) and phosphotyrosine binding (PTB and PH) domain containing proteins), proline-rich binding proteins (SH3 domain containing proteins), GTPases, phosphodiesterases, phospholipases, prolyl isomerases, proteases, Ca2+ binding proteins, cAMP binding proteins, guanyl cyclases, adenylyl cyclases, NO generating proteins, nucleotide exchange factors, and transcription factors.
  • SRC homology 2 SH2
  • PTB and PH phosphotyrosine binding
  • proline-rich binding proteins SH3 domain containing proteins
  • GTPases phosphodiesterases
  • phospholipases prolyl isomerases
  • proteases Ca2+
  • Biological activities of phosphatases according to the invention include, but are not limited to, the binding of a natural or a synthetic ligand, as well as any one of the functional activities of phosphatases known in the art.
  • Non-limiting examples of phosphatase activities include transmembrane signaling of various forms, which may involve phosphatase binding interactions and/or the exertion of an influence over signal transduction.
  • the modulators of the invention exhibit a variety of chemical structures, which can be generally grouped into mimetics of natural phosphatase ligands, and peptide and non-peptide allosteric effectors of phosphatases.
  • the invention does not restrict the sources for suitable modulators, which may be obtained from natural sources such as plant, animal or mineral extracts, or non-natural sources such as small molecule libraries, including the products of combinatorial chemical approaches to library construction, and peptide libraries.
  • Recombinant receptors are preferred for binding assay HTS because they allow for better specificity (higher relative purity), provide the ability to generate large amounts of receptor material, and can be used in a broad variety of formats (see Hodgson, Bio/Technology, 1992, 10, 973-980; each of which is incorporated herein by reference in its entirety).
  • An expressed phosphatase can be used for HTS binding assays in conjunction with its defined ligand, in this case the corresponding peptide that activates it.
  • the identified peptide is labeled with a suitable radioisotope, including, but not limited to, 125 I, 3 H, 35 S or 32 P, by methods that are well known to those skilled in the art.
  • the peptides may be labeled by well-known methods with a suitable fluorescent derivative (Baindur, et al., Drug Dev. Res., 1994, 33, 373-398; Rogers, Drug Discovery Today, 1997, 2, 156-160).
  • Radioactive ligand specifically bound to the receptor in membrane preparations made from the cell line expressing the recombinant protein can be detected in HTS assays in one of several standard ways, including filtration of the receptor-ligand complex to separate bound ligand from unbound ligand (Williams, Med. Res. Rev., 1991, 11, 147-184.; Sweetnam, et al., J. Natural Products, 1993, 56, 441-455).
  • Alternative methods include a scintillation proximity assay (SPA) or a FlashPlate format in which such separation is unnecessary (Nakayama, Cur. Opinion Drug Disc. Dev., 1998, 1, 85-91 Bossé, et al., J. Biomolecular Screening, 1998, 3, 285-292.).
  • Examples of such biological responses include, but are not limited to, the following: the ability to survive in the absence of a limiting nutrient in specifically engineered yeast cells (Pausch, Trends in Biotechnology, 1997, 15, 487-494); changes in intracellular Ca 2 + concentration as measured by fluorescent dyes (Murphy, et al., Cur. Opinion Drug Disc. Dev., 1998, 1, 192-199). Fluorescence changes can also be used to monitor ligand-induced changes in membrane potential or intracellular pH; an automated system suitable for HTS has been described for these purposes (Schroeder, et al., J. Biomolecular Screening, 1996, 1, 75-80). Assays are also available for the measurement of common second but these are not generally preferred for HTS.
  • Still other libraries of interest include peptide, protein, peptidomimetic, multiparallel synthetic collection, recombinatorial, and polypeptide libraries.
  • combinatorial chemistry and libraries created therefrom see Myers, Curr. Opin. Biotechnol. 8:701-707 (1997).
  • Identification of modulators through use of the various libraries described herein permits modification of the candidate “hit” (or “lead”) to optimize the capacity of the “hit” to modulate activity.
  • binding partners can be designed and include soluble forms of binding partners, as well as such binding partners as chimeric, or fusion, proteins.
  • assays may be used to identify specific peptide ligands of a phosphatase polypeptide, including assays that identify ligands of the target protein through measuring direct binding of test ligands to the target protein, as well as assays that identify ligands of target proteins through affinity ultrafiltration with ion spray mass spectroscopy/HPLC methods or other physical and analytical methods.
  • binding interactions are evaluated indirectly using the yeast two-hybrid system described in Fields et al., Nature, 340:245-246 (1989), and Fields et al., Trends in Genetics, 10:286-292 (1994), both of which are incorporated herein by reference.
  • the assay requires the construction of two hybrid genes encoding (1) a DNA-binding domain that is fused to a first protein and (2) an activation domain fused to a second protein.
  • the DNA-binding domain targets the first hybrid protein to the UAS of the reporter gene; however, because most proteins lack an activation domain, this DNA-binding hybrid protein does not activate transcription of the reporter gene.
  • the second hybrid protein which contains the activation domain, cannot by itself activate expression of the reporter gene because it does not bind the UAS. However, when both hybrid proteins are present, the noncovalent interaction of the first and second proteins tethers the activation domain to the UAS, activating transcription of the reporter gene.
  • this assay can be used to detect agents that interfere with the binding interaction.
  • Expression of the reporter gene is monitored as different test agents are added to the system. The presence of an inhibitory agent results in lack of a reporter signal.
  • the yeast two-hybrid assay can also be used to identify proteins that bind to the gene product.
  • a fusion polynucleotide encoding both a phosphatase polypeptide (or fragment) and a UAS binding domain i.e., a first protein
  • a large number of hybrid genes each encoding a different second protein fused to an activation domain are produced and screened in the assay.
  • the second protein is encoded by one or more members of a total-cDNA or genomic DNA fusion library, with each second protein coding region being fused to the activation domain.
  • This system is applicable to a wide variety of proteins, and it is not even necessary to know the identity or function of the second binding protein.
  • the system is highly sensitive and can detect interactions not revealed by other methods; even transient interactions may trigger transcription to produce a stable mRNA that can be repeatedly translated to yield the reporter protein.
  • the folded target protein is present to a greater extent in the presence of a test ligand which binds the target protein, than in the absence of a ligand. Binding of the ligand to the target protein can be determined by any method which distinguishes between the folded and unfolded states of the target protein. The function of the target protein need not be known in order for this assay to be performed. Virtually any agent can be assessed by this method as a test ligand, including, but not limited to, metals, polypeptides, proteins, lipids, polysaccharides, polynucleotides and small organic molecules.
  • methods of screening for compounds which modulate phosphatase activity comprise contacting test compounds with phosphatase polypeptides and assaying for the presence of a complex between the compound and the phosphatase polypeptide.
  • the ligand is typically labelled. After suitable incubation, free ligand is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of the particular compound to bind to the phosphatase polypeptide.
  • high throughput screening for compounds having suitable binding affinity to phosphatase polypeptides is employed. Briefly, large numbers of different small peptide test compounds are synthesised on a solid substrate. The peptide test compounds are contacted with the phosphatase polypeptide and washed. Bound phosphatase polypeptide is then detected by methods well known in the art. Purified polypeptides of the invention can also be coated directly onto plates for use in the aforementioned drug screening techniques. In addition, non-neutralizing antibodies can be used to capture the protein and immobilize it on the solid support.
  • Other embodiments of the invention comprise using competitive screening assays in which neutralizing antibodies capable of binding a polypeptide of the invention specifically compete with a test compound for binding to the polypeptide.
  • the antibodies can be used to detect the presence of any peptide that shares one or more antigenic determinants with a phosphatase polypeptide.
  • Radiolabeled competitive binding studies are described in A. H. Lin et al. Antimicrobial Agents and Chemotherapy, 1997, vol. 41, no. 10. pp. 2127-2131, the disclosure of which is incorporated herein by reference in its entirety.
  • the invention provides methods for treating a disease by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
  • these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic bowel
  • the invention provides methods for treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
  • these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic bowel
  • the invention also features methods of treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
  • the invention also features methods of treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
  • the disease is selected from the group consisting of immune-related diseases and disorders, cardiovascular disease, and cancer.
  • preventing refers to decreasing the probability that an organism contracts or develops an abnormal condition.
  • treating refers to having a therapeutic effect and at least partially alleviating or abrogating an abnormal condition in the organism.
  • abnormal condition refers to a function in the cells or tissues of an organism that deviates from their normal functions in that organism.
  • An abnormal condition can relate to cell proliferation, cell differentiation, or cell survival.
  • Abnormal cell proliferative conditions include cancers such as fibrotic and mesangial disorders, abnormal angiogenesis and vasculogenesis, wound healing, psoriasis, diabetes mellitus, and inflammation.
  • Abnormal cell survival conditions relate to conditions in which programmed cell death (apoptosis) pathways are activated or abrogated.
  • a number of protein phosphatases are associated with the apoptosis pathways. Aberrations in the function of any one of the protein phosphatases could lead to cell immortality or premature cell death.
  • administering relates to a method of incorporating a compound into cells or tissues of an organism.
  • the abnormal condition can be prevented or treated when the cells or tissues of the organism exist within the organism or outside of the organism.
  • Cells existing outside the organism can be maintained or grown in cell culture dishes.
  • many techniques exist in the art to administer compounds including (but not limited to) oral, parenteral, dermal, injection, and aerosol applications.
  • multiple techniques exist in the art to administer the compounds including (but not limited to) cell microinjection techniques, transformation techniques, and carrier techniques.
  • the abnormal condition can also be prevented or treated by administering a compound to a group of cells having an aberration in a signal transduction pathway to an organism.
  • the effect of administering a compound on organism function can then be monitored.
  • the organism is preferably a mouse, rat, rabbit, guinea pig, or goat, more preferably a monkey or ape, and most preferably a human.
  • the invention features methods for detection of a phosphatase polypeptide in a sample as a diagnostic tool for diseases or disorders, wherein the method comprises the steps of: (a) contacting the sample with a nucleic acid probe which hybridizes under hybridization assay conditions to a nucleic acid target region of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, said probe comprising the nucleic acid sequence encoding the polypeptide, fragments thereof, and the complements of the sequences and fragments; and (b) detecting the presence or amount of the probe:target region hybrid as an indication of the disease.
  • the disease or disorder is selected from the group consisting of rheumatoid arthritis, arteriosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure, oxidative stress-related neurodegenerative disorders, and cancer.
  • the phosphatase “target region” is the nucleotide base sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or the corresponding full-length sequences, a functional derivative thereof, or a fragment thereof to which the nucleic acid probe will specifically hybridize. Specific hybridization indicates that in the presence of other nucleic acids the probe only hybridizes detectably with the phosphatase of the invention's target region. Putative target regions can be identified by methods well known in the art consisting of alignment and comparison of the most closely related sequences in the database.
  • the nucleic acid probe hybridizes to a phosphatase target region encoding at least 6, 12, 75, 90, 105, 120, 150, 200, 250, 300 or 350 contiguous amino acids of a sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or the corresponding full-length amino acid sequence, or a functional derivative thereof.
  • Hybridization conditions should be such that hybridization occurs only with the phosphatase genes in the presence of other nucleic acid molecules. Under stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having more than 1 or 2 mismatches out of 20 contiguous nucleotides. Such conditions are defined, above.
  • RNA can be the detectable presence of phosphatase RNA in cells, since in some normal cells there is no basal expression of phosphatase RNA. In other normal cells, a basal level of expression of phosphatase exists, therefore in these cases amplification is the detection of at least 1-2-fold, and preferably more, phosphatase RNA, compared to the basal level.
  • the invention features a method for detection of a phosphatase polypeptide in a sample as a diagnostic tool for a disease or disorder, wherein the method comprises: (a) comparing a nucleic acid target region encoding the phosphatase polypeptide in a sample, where the phosphatase polypeptide has an amino acid sequence selected from the group consisting those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or one or more fragments thereof, with a control nucleic acid target region encoding the phosphatase polypeptide, or one or more fragments thereof; and (b) detecting differences in sequence or amount between the target region and the control target region, as an indication of the disease or disorder.
  • FIGS. 1 A-H show the nucleotide sequences for human protein phosphatases (SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12).
  • accession number for example, ss1581624 for SGP187
  • dbSNP the database of single nucleotide polymorphisms maintained at NCBI (http://www.ncbi.nh.nih.gov/SNP/index.html).
  • the accession number for SNP can be used to retrieve the full SNP-containing sequence from this site.
  • Candidate SNPs without a dbSNP accession number were identified by inspection of Blastn outputs of the patent sequences vs cDNA and genomic databases, as shown in Table 7 and Table 8, respectively, in Example 1.
  • the present invention additionally provides nucleic acid probes an uses therefor.
  • a nucleic acid probe of the present invention may be used to probe an appropriate chromosomal or cDNA library by usual hybridization methods to obtain other nucleic acid molecules of the present invention.
  • a chromosomal DNA or cDNA library may be prepared from appropriate cells according to recognized methods in the art (cf. “Molecular Cloning: A Laboratory Manual”, second edition, Cold Spring Harbor Laboratory, Sambrool, Fritsch, & Maniatis, eds., 1989).
  • nucleic acid probes having nucleotide sequences which correspond to N-terminal and C-terminal portions of the amino acid sequence of the polypeptide of interest.
  • the synthesized nucleic acid probes may be used as primers in a polymerase chain reaction (PCR) carried out in accordance with recognized PCR techniques, essentially according to PCR Protocols, “A Guide to Methods and Applications”, Academic Press, Michael, et al., eds., 1990, utilizing the appropriate chromosomal or cDNA library to obtain the fragment of the present invention.
  • PCR polymerase chain reaction
  • hybridization probes of the present invention can be labeled by standard labeling techniques such as with a radiolabel, enzyme label, fluorescent label, biotin-avidin label, chemiluminescence, and the like. After hybridization, the probes may be visualized using known methods.
  • the nucleic acid probes of the present invention include RNA, as well as DNA probes, such probes being generated using techniques known in the art.
  • the nucleic acid probe may be immobilized on a solid support.
  • solid supports include, but are not limited to, plastics such as polycarbonate, complex carbohydrates such as agarose and sepharose, and acrylic resins, such as polyacrylamide and latex beads. Techniques for coupling nucleic acid probes to such solid supports are well known in the art.
  • One method of detecting the presence of nucleic acids of the invention in a sample comprises (a) contacting said sample with the above-described nucleic acid probe under conditions such that hybridization occurs, and (b) detecting the presence of said probe bound to said nucleic acid molecule.
  • One skilled in the art would select the nucleic acid probe according to techniques known in the art as described above. Samples to be tested include but should not be limited to RNA samples of human tissue.
  • a kit for detecting the presence of nucleic acids of the invention in a sample comprises at least one container means having disposed therein the above-described nucleic acid probe.
  • the kit may further comprise other containers comprising one or more of the following: wash reagents and reagents capable of detecting the presence of bound nucleic acid probe.
  • detection reagents include, but are not limited to radiolabelled probes, enzymatic labeled probes (horseradish peroxidase, alkaline phosphatase), and affinity labeled probes (biotin, avidin, or steptavidin).
  • the kit further comprises instructions for use.
  • a compartmentalized kit includes any kit in which reagents are contained in separate containers.
  • Such containers include small glass containers, plastic containers or strips of plastic or paper.
  • Such containers allow the efficient transfer of reagents from one compartment to another compartment such that the samples and reagents are not cross-contaminated and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another.
  • Such containers will include a container which will accept the test sample, a container which contains the probe or primers used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, and the like), and containers which contain the reagents used to detect the hybridized probe, bound antibody, amplified product, or the like.
  • wash reagents such as phosphate buffered saline, Tris-buffers, and the like
  • a classification of the protein class and family to which it belongs a summary of non-catalytic protein motifs, as well as a chromosomal location. This information is useful in determing function, regulation and/or therapeutic utility for each of the proteins. Amplification of chromosomal region can be associated with various cancers.
  • the phosphatase classification and protein domains often reflect pathways, cellular roles, or mechanisms of up- or down-stream regulation. Also disease-relevant genes often occur in families of related genes. For example, if one member of a phosphatase family functions as an oncogene, a tumor suppressor, or has been found to be disrupted in an immune, neurologic, cardiovascular, or metabolic disorder, frequently other family members may play a related role.
  • Chromosomal location can identify candidate targets for a tumor amplicon or a tumor-suppressor locus. Summaries of prevalent tumor amplicons are available in the literature, and can identify tumor types to experimentally be confirmed to contain amplified copies of a phosphatase gene which localizes to an adjacent region.
  • polypeptides described in the present invention may belong to one of the following groups: (1) dual-specificity group of protein phosphatases (DSP); (2) serine-threonine phosphatases (STP); or (3) protein tyrosine phosphatases (PTP).
  • DSP dual-specificity group of protein phosphatases
  • STP serine-threonine phosphatases
  • PTP protein tyrosine phosphatases
  • Novel MKP-like phosphatases in this application include SGP006 (SEQ ID NO: 1), SGP002 (SEQ ID NO: 2), SGP001 (SEQ ID NO: 3), SGP018 (SEQ ID NO: 4), SGP003 (SEQ ID NO: 5), SGP014 (SEQ ID NO: 6), SGP060 (SEQ ID NO: 7), and SGP008 (SEQ ID NO: 8), which are disclosed in greater detail in the Tables 1-6 and Example 2, for example.
  • MKP phosphatases are capable of inactivating, through a dephosphorylation reaction, kinases that participate in the MAPK pathways.
  • the ERK (extracellular signal-regulated kinase), JNK/SAPK (c-Jun N-terminal kinase/stress-activated protein kinase) and p38 MAP kinase pathways mediate the signal transduction events that are responsible for cell division, differentiation or apoptosis in response to extracellular ligands (Cobb M H, Prog Biophys Mol Biol. 1999;71(3-4):479-500).
  • MKP-1 Over-expression of MKP-1 in prostate tumor cell lines confers resistance to Fas ligand-induced apoptosis (Srikanth, S. et al. (1999) Mol. Cell. Biochem. 199: 169-178) and it has also been suggested that MKP-1 may contribute to the inhibition of apoptosis resulting in androgen-independent growth. MKP-1 may also inhibit the induction of apoptosis that is produced by anti-neoplastic agents such as cisplatin and camptothecin (Sanchez-Perez, I et al. (2000) Oncogene 19: 5142-5152; Costa-Pereira, A. P. et al. (2000) Br. J. Cancer 82: 1827-1834).
  • anti-neoplastic agents such as cisplatin and camptothecin
  • ERK-1 and ERK-2 by MAPK phosphatases may also be responsible for suppressing angiogenic vascular endothelial cell proliferation by angiostatin Redlitz, A. et al. (1999 J. Vasc. Res 36:28-34).
  • novel MPK family phosphatases presented in this filing contribute to a growing list of phosphatases that appear to have as their primary function negative feedback regulation of MAPK signal transduction. Since there is precedence for selectivity in the mechanism of action at the level of substrate recognition, subcellular localization and tissue distribution among the known MPK's, the novel MPK's described may display similar selectivity. The novel MPK's may also play a role in suppressing apoptosis by blocking the JNK/SAPK pathway during pathological hypoxia such as that occurring in angiogenic tumors. The development of specific phosphatase inhibitors that target the anti-apoptotic MKP's may prove valuable as an approach to cancer therapy.
  • SGP012 SEQ ID NO: 11
  • SGP024 SEQ ID NO: 12
  • SGP012 is closely related to murine OST-PTP, also called PTP-ESP.
  • Osteotesticular PTP (OST-PTP) is a putative receptor protein tyrosine phosphatase that possesses 10 fibronectin type m repeats, a potential membrane-spanning region and an intracellular domain consisting of two tandem catalytic domains. The expression pattern is highly restricted and is detectable primarily in bone and testis (Mauro et al. J. Biol Chem 1996 269:30659-67). The ligand for OST-PTP is not known but the structure of the extracellular domain suggests that cell-cell interactions may be involved. Importantly, the human ortholog has not yet been cloned.
  • OST-PTP activity is responsible for the observed increases in bone-forming activities in osteoblast cultures. Therefore targeting of OST-PTP activity could provide treatments for osteoporosis, non-healing fractures, and other disorders of bone metabolism.
  • SGP024 represents a partial PTPT catalytic domain related to PTP-delta.
  • SGP039 SEQ ID NO: 9
  • SGP040 SEQ ID NO: 10
  • the Serine-threonine phosphatases can be divided into four major classes represented by PP1, PP2A, PP2B, and PP2C.
  • PP2a is found associated with multiple regulatory subunits and its inactivation leads to transformation by viral components such as small T antigen. Mutations in one of the regulatory subunits have been associated with colorectal cancers consistent with a role as a tumor suppressor (Talcagi et al. Gut 2000 47:268-71. Recently, PP2a has also been implicated in activation of T lymphocytes (Chuang et al. Immunity 2000 13:313-22).
  • PP1 has been implicated in a variety of cellular functions including response to hypoxia, apoptosis and cytokinesis (Taylor et al., PNAS 2000 97:12091-96, Aylion et al. EMBO J 2000 19 2237-46, Orr et al., Infect. Immun. 2000 68:1350-58). Finally, studies in diabetic rats showed decreased PP1 activity and elevated PP2A activity compared to controls (Begum and Ragolia Metabolism 1998 47:54-62). Because of the diversity of regulatory subunits that affect the activity of serine-threonine phosphatases, biological function of novel members are difficult to predict. However, the studies suggest potential involvement in a variety of diseases including tumorigenesis, inflammatory diseases, and metabolic diseases.
  • the invention provides methods for detecting a polypeptide in a sample as a diagnostic tool for diseases or disorders, wherein the method comprises the steps of: (a) contacting the sample with a nucleic acid probe which hybridizes under hybridization assay conditions to a nucleic acid target region of a polypeptide selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17; SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, said probe comprising the nucleic acid sequence encoding the polypeptide, fragments thereof, and the complements of the sequences and fragments; and (b) detecting the presence or amount of the probe:target region hybrid as an indication of the disease.
  • the disease or disorder is selected from the group consisting of rheumatoid arthritis, atherosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure, oxidative stress-related neurodegenerative disorders, metabolic disorder including diabetes, reproductive disorders including infertility, and cancer.
  • Hybridization conditions should be such that hybridization occurs only with the genes in the presence of other nucleic acid molecules. Under stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having 1 or 2 mismatches out of 20 contiguous nucleotides. Such conditions are defined herein.
  • the diseases for which detection of genes in a sample could be diagnostic include diseases in which nucleic acid (DNA and/or RNA) is amplified in comparison to normal cells.
  • amplification is meant increased numbers of DNA or RNA in a cell compared with normal cells.
  • RNA can be the detectable presence of RNA in cells, since in some normal cells there is no basal expression of RNA. In other normal cells, a basal level of expression exists, therefore in these cases amplification is the detection of at least 1-2-fold, and preferably more, compared to the basal level.
  • the diseases that could be diagnosed by detection of nucleic acid in a sample preferably include cancers.
  • the test samples suitable for nucleic acid probing methods of the present invention include, for example, cells or nucleic acid extracts of cells, or biological fluids.
  • the samples used in the above-described methods will vary based on the assay format, the detection method and the nature of the tissues, cells or extracts to be assayed. Methods for preparing nucleic acid extracts of cells are well known in the art and can be readily adapted in order to obtain a sample that is compatible with the method utilized.
  • the present invention relates to an antibody having binding affinity to a phosphatase of the invention.
  • the polypeptide may have the amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or a functional derivative thereof, or at least 9 contiguous amino acids thereof (preferably, at least 20, 30, 35, or 40 contiguous amino acids thereof).
  • the present invention also relates to an antibody having specific binding affinity to a phosphatase of the invention.
  • an antibody may be isolated by comparing its binding affinity to a phosphatase of the invention with its binding affinity to other polypeptides.
  • Those which bind selectively to a phosphatase of the invention would be chosen for use in methods requiring a distinction between a phosphatase of the invention and other polypeptides.
  • Such methods could include, but should not be limited to, the analysis of altered phosphatase expression in tissue containing other polypeptides.
  • the phosphatases of the present invention can be used in a variety of procedures and methods, such as for the generation of antibodies, for use in identifying pharmaceutical compositions, and for studying DNA/protein interaction.
  • the present invention also relates to a hybridoma which produces the above-described monoclonal antibody, or binding fragment thereof.
  • a hybridoma is an immortalized cell line which is capable of secreting a specific monoclonal antibody.
  • the polypeptide may be modified or administered in an adjuvant in order to increase the peptide antigenicity.
  • Methods of increasing the antigenicity of a polypeptide are well known in the art. Such procedures include coupling the antigen with a heterologous protein (such as globulin or ⁇ -galactosidase) or through the inclusion of an adjuvant during immunization.
  • a heterologous protein such as globulin or ⁇ -galactosidase
  • spleen cells from the immunized animals are removed, fused with myeloma cells, such as SP2/0-Ag14 myeloma cells, and allowed to become monoclonal antibody producing hybridoma cells.
  • myeloma cells such as SP2/0-Ag14 myeloma cells
  • Any one of a number of methods well known in the art can be used to identify the hybridoma cell which produces an antibody with the desired characteristics. These include screening the hybridomas with an ELISA assay, western blot analysis, or radioimmunoassay Lutz et al., Exp. Cell Res. 175:109-124, 1988).
  • Hybridomas secreting the desired antibodies are cloned and the class and subclass are determined using procedures known in the art (Campbell, “Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology”, supra, 1984).
  • Anti-peptide peptides can be generated by replacing the basic amino acid residues found in the peptide sequences of the phosphatases of the invention with acidic residues, while maintaining hydrophobic and uncharged polar groups. For example, lysine, arginine, and/or histidine residues are replaced with aspartic acid or glutamic acid and glutamic acid residues are replaced by lysine, arginine or histidine.
  • Conditions for incubating an antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the antibody used in the assay.
  • immunological assay formats such as radioimmunoassays, enzyme-linked immunosorbent assays, diffusion-based Ouchterlony, or rocket immunofluorescent assays
  • Examples of such assays can be found in Chard (“An Introduction to Radioimmunoassay and Related Techniques” Elsevier Science Publishers, Amsterdam, The Netherlands, 1986), Bullock et al.
  • the immunological assay test samples of the present invention include cells, protein or membrane extracts of cells, or biological fluids such as blood, serum, plasma, or urine.
  • the test samples used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed. Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can readily be adapted in order to obtain a sample which is testable with the system utilized.
  • detection reagents include, but are not limited to, labeled secondary antibodies, or in the alternative, if the primary antibody is labeled, the chromophoric, enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody.
  • the compartmentalized kit may be as described above for nucleic acid probe kits.
  • the antibodies described in the present invention can readily be incorporated into one of the established kit formats which are well known in the art.
  • the present invention also relates to a method of detecting a compound capable of binding to a phosphatase of the invention comprising incubating the compound with a phosphatase of the invention and detecting the presence of the compound bound to the phosphatase.
  • the compound may be present within a complex mixture, for example, serum, body fluid, or cell extracts.
  • the present invention also relates to a method of detecting an agonist or antagonist of phosphatase activity or phosphatase binding partner activity comprising incubating cells that produce a phosphatase of the invention in the presence of a compound and detecting changes in the level of phosphatase activity or phosphatase binding partner activity.
  • the compounds thus identified would produce a change in activity indicative of the presence of the compound.
  • the compound may be present within a complex mixture, for example, serum, body fluid, or cell extracts. Once the compound is identified it can be isolated using techniques well known in the art.
  • the invention additionally provides methods for treating a disease or abnormal condition by administering to a patient in need of such treatment a substance that modulates the activity of a polypeptide selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, a functional derivative thereof, and a fragment thereof.
  • a polypeptide selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, a functional derivative thereof, and a fragment thereof.
  • the disease is selected from the group consisting of rheumatoid arthritis, atherosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure; oxidative stress-related neurodegenerative disorders, metabolic and reproductive disorders, and cancer.
  • Substances useful for treatment of disorders or diseases preferably show positive results in one or more assays for an activity corresponding to treatment of the disease or disorder in question
  • Substances that modulate the activity of the polypeptides preferably include, but are not limited to, antisense oligonucleotides and inhibitors of protein phosphatases.
  • preventing refers to decreasing the probability that an organism contracts or develops an abnormal condition.
  • a therapeutic effect refers to the inhibition or activation factors causing or contributing to the abnormal condition.
  • a therapeutic effect relieves to some extent one or more of the symptoms of the abnormal condition.
  • a therapeutic effect can refer to one or more of the following: (a) an increase in the proliferation, growth, and/or differentiation of cells; (b) inhibition (, slowing or stopping) of cell death; (c) inhibition of degeneration; (d) relieving to some extent one or more of the symptoms associated with the abnormal condition; and (e) enhancing the function of the affected population of cells.
  • Compounds demonstrating efficacy against abnormal conditions can be identified as described herein.
  • abnormal condition refers to a function in the cells or tissues of an organism that deviates from their normal functions in that organism.
  • An abnormal condition can relate to cell proliferation, cell differentiation or cell survival.
  • An abnormal condition may also include irregularities in cell cycle progression, i.e., irregularities in normal cell cycle progression through mitosis and meiosis.
  • Abnormal differentiation conditions include, but are not limited to, neurodegenerative disorders, slow wound healing rates, and slow tissue grafting healing rates.
  • administering relates to a method of incorporating a compound into cells or tissues of an organism.
  • the abnormal condition can be prevented or treated when the cells or tissues of the organism exist within the organism or outside of the organism.
  • Cells existing outside the organism can be maintained or grown in cell culture dishes.
  • many techniques exist in the art to administer compounds including (but not limited to) oral, parenteral, dermal, injection, and aerosol applications.
  • multiple techniques exist in the art to administer the compounds including (but not limited to) cell microinjection techniques, transformation techniques and carrier techniques.
  • the abnormal condition can also be prevented or treated by administering a compound to a group of cells having an aberration in a signal transduction pathway to an organism.
  • the effect of administering a compound on organism function can then be monitored.
  • the organism is preferably a mouse, rat, rabbit, guinea pig or goat, more preferably a monkey or ape, and most preferably a human.
  • the present invention also encompasses a method of agonizing (stimulating) or antagonizing phosphatase associated activity in a mammal comprising administering to said mammal an agonist or antagonist to an amino acid sequence selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, a functional derivative thereof, and a fragment thereof in an amount sufficient to effect said agonism or antagonism.
  • the present application also contemplates a method of treating diseases in a mammal with an agonist or antagonist of the activity of one of the above mentioned polypeptides of the invention comprising administering the agonist or antagonist to a mammal in an amount sufficient to agonize or antagonize a phosphatase-associated function.
  • the present invention provides methods for treating pathologies, including breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer.
  • phosphatase gene sequences used include: SEQ ID NO4, SEQ ID NO: 5, and SEQ ID NO: 7.
  • samples from normal tissues, tumor tissues, various cell lines, and P53 wild type and mutant were used to make the expression array.
  • the relative gene expression levels of the tested phosphatase genes in various tissue sources and cell lines were quantitated by measuring Syber Green I staining of hybridized signals. The numerical readings recorded in the table were normalized to the hybridization result from ds cDNA or undenatured probes, after subtracting the background counts.
  • the relevant expression levels in Table 5 constitutes expression profiles of the phosphatase genes of interest in various tissue sources.
  • Such expression profile data guides application of the treatment regime according to the present invention.
  • “M14” cell line (a malignant melanoma) in Table 5 the levels of expression of SEQ ID NO: 4 is zero.
  • the level of expression of SEQ ID NO: 7 (58) is low to marginal.
  • the level of expression of SEQ ID NO: 5 (2,528) is significantly higher.
  • Such horizontal comparison reveals that the phosphatase gene encoded by SEQ ID NO: 5 is implicated in melanoma.
  • the diagnostic method of the present invention may be used to test for breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer.
  • the data obtained from the microarray data also can be used to diagnose a patient who may be suffering from a particular pathology.
  • a method of diagnosing the cancer condition connected to melanoma, according to the present invention is, therefore, to contact a test sample, which may be collected from a patient, with a nucleotide probe which is capable of hybridizing to the nucleic acid sequence which encodes the protein represented by SEQ ID NO: 17; and then to detect the presence of the hybridized probe:target pairs and to quantify the level of such hybridization as an indication of the cancer condition connected to neuroblastoma.
  • the expression analysis according to the preferred embodiment of this invention thus, confers specificity and effectiveness to the diagnostic method disclosed.
  • comparison and correlation analysis may be carried out based on expression data generated in the way similar to that described here; they also necessarily fall in the scope of the present invention. Inferences derived from those comparison and correlation analysis may similarly be used in substantiating the diagnostic method according to this invention.
  • One scenario to be noted is when pairs of samples of normal tissues and diseased tissues are used to make the expression arrays, the data generated will specifically demonstrate which phosphatase genes are differentially expressed in certain diseased conditions, therefore may serve as diagnostic markers used in the aforementioned diagnostic method.
  • This method also may be used for diagnosing a wide range of diseases, including cancer, pathophysiological hypoxia, cardiovascular disorders, Papillon-Lefevre syndrome, Cowden disease, ectordermal dysplasia, Moebius syndrome, Bjomstad syndrome, Bannayan Zonana syndrome, schizophrenia and hamartomas. Of particular importance is diagnosis of various type of cancers.
  • this particular method may similarly be used to test for breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer.
  • a target region can be any particular region of interest in a phosphatase gene, such as an upstream regulatory region. Variations of sequence in an upstream regulatory region in a family of phosphatase often have functional implications some of which may be significant in bringing about certain diseased conditions. Changes of the amount of a target region, e.g., changes of number of copies of a regulatory region such as a receptor-binding site, in certain phosphatase genes, may also represent mechanisms of functional differentiation and hence may be connected to certain diseased states. Detection of such differences in sequence and amount of a target region compared to a control region therefore may effectively lead to detection of a diseased condition.
  • microarray studies may be used to identify the potential connections between a diseased condition and variations of a target region among a set of phosphatase genes.
  • nucleic acid probes may be made that correspond to a given target region and a control region, respectively, of a phosphatase gene of interest.
  • Samples from normal and diseased tissues are used to make microarray as discussed, supra, and in Example 3. Hybridization of these probes to the array so made will yield comparative profiles of the region of interest in the normal and diseased condition, and thus may derive a definition of differences of the target region and control region that is characterized of the disease in question.
  • Such definition may serve as an indication of the diseased condition as used in the second-mentioned diagnostic method according to the present invention. It should be appreciated that many equivalent or similar methods may be used in carrying out the diagnosis according to this method which would become apparent to the skilled person in the art based on the example provided here, and therefore, they are covered in the scope of this invention.
  • indolinone compounds form classes of acid resistant and membrane permeable organic molecules.
  • WO 96/22976 published Aug. 1, 1996 by Ballinari et al. describes hydrosoluble indolinone compounds that harbor tetralin, naphthalene, quinoline, and indole substituents fused to the oxindole ring. These bicyclic substituents are in turn substituted with polar moieties including hydroxylated alkyl, phosphate, and ether moieties.
  • U.S. patent application Ser. No. 08/702,232 filed Aug. 23, 1996, entitled “Indolinone Combinatorial Libraries and Related Products and Methods for the Treatment of Disease” by Tang et al. (U.S. Ser.
  • Other examples of substances capable of modulating phosphatase activity include, but are not limited to, tyrphostins, quinazolines, quinoxolines, and quinolines.
  • the quinazolines, tyrphostins, quinolines, and quinoxolines referred to above include well known compounds such as those described in the literature.
  • representative publications describing quinazolines include Barker et al., EPO Publication No. 0 520 722 Al; Jones et al., U.S. Pat. No. 4,447,608; Kabbe et al., U.S. Pat. No. 4,757,072; Kaul and Vougioukas, U.S. Pat. No.
  • oxindolinones such as those described in U.S. patent application Ser. No. 08/702,232 filed Aug. 23, 1996, incorporated herein by reference in its entirety, including any drawings.
  • the present invention also relates to a recombinant DNA molecule comprising, 5′ to 3′, a promoter effective to initiate transcription in a host cell and the above-described nucleic acid molecules.
  • the present invention relates to a recombinant DNA molecule comprising a vector and an above-described nucleic acid molecule.
  • the present invention also relates to a nucleic acid molecule comprising a transcriptional region functional in a cell, a sequence complementary to an RNA sequence encoding an amino acid sequence corresponding to the above-described polypeptide, and a transcriptional termination region functional in said cell.
  • the above-described molecules may be isolated and/or purified DNA molecules.
  • a nucleic acid molecule such as DNA
  • a nucleic acid molecule is said to be “capable of expressing” a polypeptide if it contains nucleotide sequences which contain transcriptional and translational regulatory information and such sequences are “operably linked” to nucleotide sequences which encode the polypeptide.
  • An operable linkage is a linkage in which the regulatory DNA sequences and the DNA sequence sought to be expressed are connected in such a way as to permit gene sequence expression.
  • regulatory regions needed for gene sequence expression may vary from organism to organism, but shall in general include a promoter region which, in prokaryotes, contains both the promoter (which directs the initiation of RNA transcription) as well as the DNA sequences which, when transcribed into RNA, will signal synthesis initiation.
  • promoter region which, in prokaryotes, contains both the promoter (which directs the initiation of RNA transcription) as well as the DNA sequences which, when transcribed into RNA, will signal synthesis initiation.
  • Such regions will normally include those 5′-non-coding sequences involved with initiation of transcription and translation, such as the TATA box, capping sequence, CAAT sequence, and the like.
  • the non-coding region 3′ to the sequence encoding a phosphatase of the invention may be obtained by the above-described methods. This region may be retained for its transcriptional termination regulatory sequences, such as termination and polyadenylation. Thus, by retaining the 3′-region naturally contiguous to the DNA sequence encoding a phosphatase of the invention, the transcriptional termination signals may be provided. Where the transcriptional termination signals are not satisfactorily functional in the expression host cell, then a 3′ region functional in the host cell may be substituted.
  • Two DNA sequences are said to be operably linked if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the promoter region sequence to direct the transcription of a gene sequence encoding a phosphatase of the invention, or (3) interfere with the ability of the gene sequence of a phosphatase of the invention to be transcribed by the promoter region sequence.
  • a promoter region would be operably linked to a DNA sequence if the promoter were capable of effecting transcription of that DNA sequence.
  • the present invention encompasses the expression of a gene encoding a phosphatase of the invention (or a functional derivative thereof) in either prokaryotic or eukaryotic cells.
  • Prokaryotic hosts are, generally, very efficient and convenient for the production of recombinant proteins and are, therefore, one type of preferred expression system for phosphatases of the invention.
  • Prokaryotes most frequently are represented by various strains of E. coli. However, other microbial strains may also be used, including other bacterial strains.
  • plasmid vectors that contain replication sites and control sequences derived from a species compatible with the host may be used.
  • suitable plasmid vectors may include pBR322, pUC118, pUC119 and the like;
  • suitable phage or bacteriophage vectors may include ⁇ gt10, ⁇ gt11 and the like; and
  • suitable virus vectors may include pMAM-neo, pKRC and the like.
  • the selected vector of the present invention has the capacity to replicate in the selected host cell.
  • Recognized prokaryotic hosts include bacteria such as E. coli, Bacillus, Streptomyces, Pseudomonas, Salmonella, Seiratia, and the like. However, under such conditions, the polypeptide will not be glycosylated.
  • the prokaryotic host must be compatible with the replicon and control sequences in the expression plasmid.
  • a phosphatase of the invention (or a functional derivative thereof) in a prokaryotic cell, it is necessary to operably link the sequence encoding the phosphatase of the invention to a functional prokaryotic promoter.
  • promoters may be either constitutive or, more preferably, regulatable (i.e., inducible or derepressible).
  • constitutive promoters include the int promoter of bacteriophage ⁇ , the bla promoter of the ⁇ -lactamase gene sequence of pBR322, and the cat promoter of the chloramphenicol acetyl transferase gene sequence of pPR325, and the like.
  • inducible prokaryotic promoters examples include the major right and left promoters of bacteriophage ⁇ (P L and P R ), the tip, recA, ⁇ acZ, ⁇ acI, and gal promoters of E. coli, the ⁇ -amylase (Ulmanen et al., J. Bacteriol. 162:176-182, 1985) and the ⁇ -28-specific promoters of B.
  • progeny Proper expression in a prokaryotic cell also requires the presence of a ribosome-binding site upstream of the gene sequence-encoding sequence.
  • ribosome-binding sites are disclosed, for example, by Gold et al. (Ann. Rev. Microbiol. 35:365-404, 1981).
  • the selection of control sequences, expression vectors, transformation methods, and the like, are dependent on the type of host cell used to express the gene.
  • “cell”, “cell line”, and “cell culture” may be used interchangeably and all such designations include progeny.
  • the words “transformants” or “transformed cells” include the primary subject cell and cultures derived therefrom, without regard to the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. However, as defined, mutant progeny have the same functionality as that of the originally transformed cell.
  • Host cells which may be used in the expression systems of the present invention are not strictly limited, provided that they are suitable for use in the expression of the phosphatase polypeptide of interest. Suitable hosts may often include eukaryotic cells. Preferred eukaryotic hosts include, for example, yeast, fungi, insect cells, mammalian cells either in vivo, or in tissue culture. Mammalian cells which may be useful as hosts include HeLa cells, cells of fibroblast origin such as VERO or CHO-K1, or cells of lymphoid origin and their derivatives. Preferred mammalian host cells include SP2/0 and J558L, as well as neuroblastoma cell lines such as IMR 332, which may provide better capacities for correct post-translational processing.
  • Any of a series of yeast expression systems can be utilized which incorporate promoter and termination elements from the actively expressed sequences coding for glycolytic enzymes that are produced in large quantities when yeast are grown in mediums rich in glucose.
  • Known glycolytic gene sequences can also provide very efficient transcriptional control signals.
  • Yeast provides substantial advantages in that it can also carry outpost-translational modifications.
  • Yeast recognizes leader sequences on cloned mammalian genes and secretes peptides bearing leader sequences (i.e., pre-peptides).
  • Several possible vector systems are available for the expression of phosphatases of the invention in a mammalian host.
  • eukaryotic regulatory regions Such regions will, in general, include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence Namer et al., J. Mol. Appl. Gen. 1:273-288, 1982); the TK promoter of Herpes virus (McKnight, Cell 31:355-365, 1982); the SV40 early promoter (Benoist et al., Nature (London) 290:304-31, 1981); and the yeast gal4 gene sequence promoter (Johnston et al., Proc. Natl. Acad. Sci. (USA) 79:6971-6975, 1982; Silver et al., Proc. Natl. Acad. Sci. (USA) 81:5951-5955, 1984).
  • a nucleic acid molecule encoding a phosphatase of the invention and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a nonreplicating DNA or RNA molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule. Since such molecules are incapable of autonomous replication, the expression of the gene may occur through the transient expression of the introduced sequence. Alternatively, permanent expression may occur through the integration of the introduced DNA sequence into the host chromosome.
  • DNA can be injected into the pronucleus of a fertilized egg before fusion of the male and female pronuclei, or injected into the nucleus of an embryonic cell (e.g., the nucleus of a two-cell embryo) following the initiation of cell division (Brinster et al., Proc. Nat. Acad. Sci. USA 82:4438-4442, 1985).
  • Embryos can be infected with viruses, especially retroviruses, modified to carry inorganic-ion receptor nucleotide sequences of the invention.
  • Pluripotent stem cells derived from the inner cell mass of the embryo and stabilized in culture can be manipulated in culture to incorporate nucleotide sequences of the invention.
  • a transgenic animal can be produced from such cells through implantation into a blastocyst that is implanted into a foster mother and allowed to come to term. Animals suitable for transgenic experiments can be obtained from standard commercial sources such as Charles River (Wilmington, Mass.), Taconic (Germantown, N.Y.), Harlan Sprague Dawley (Indianapolis, Ind.), etc.
  • transgenic mouse female mice are induced to superovulate. Females are placed with males, and the mated females are sacrificed by CO 2 asphyxiation or cervical dislocation and embryos are recovered from excised oviducts. Surrounding cumulus cells are removed. Pronuclear embryos are then washed and stored until the time of injection. Randomly cycling adult female mice are paired with vasectomized males. Recipient females are mated at the same time as donor females. Embryos then are transferred surgically. The procedure for generating transgenic rats is similar to that of mice (Hammer et al., Cell 63:1099-1112, 1990).
  • a clone containing the sequence(s) of the invention is co-transfected with a gene encoding resistance.
  • the gene encoding neomycin resistance is physically linked to the sequence(s) of the invention.
  • Transfection and isolation of desired clones are carried out by any one of several methods well known to those of ordinary skill in the art (E. J. Robertson, supra).
  • the final phase of the procedure is to inject targeted ES cells into blastocysts and to transfer the blastocysts into pseudopregnant females.
  • the resulting chimeric animals are bred and the offspring are analyzed by Southern blotting to identify individuals that carry the transgene.
  • Procedures for the production of non-rodent mammals and other animals have been discussed by others (Houdebine and Chourrout, supra; Pursel et al., Science 244:1281-1288, 1989; and Simms et al., Bio/Technology 6:179-183, 1988).
  • a “transgenic animal” is an animal having cells that contain DNA which has been artificially inserted into a cell, which DNA becomes part of the genome of the animal which develops from that cell.
  • Preferred transgenic animals are primates, mice, rats, cows, pigs, horses, goats, sheep, dogs and cats.
  • the transgenic DNA may encode human kinases. Native expression in an animal may be reduced by providing an amount of antisense RNA or DNA effective to reduce expression of the receptor.
  • Phosphatases or their genetic sequences will also be useful in gene therapy (reviewed in Miller, Nature 357:455-460, 1992). Miller states that advances have resulted in practical approaches to human gene therapy that have demonstrated positive initial results. The basic science of gene therapy is described in Mulligan ( Science 260:926-931, 1993).
  • the gene therapy may involve the use of an adenovirus containing phosphatase cDNA targeted to a tumor, systemic phosphatase increase by implantation of engineered cells, injection with phosphatase-encoding virus, or injection of naked phosphatase DNA into appropriate tissues.
  • Target cell populations may be modified by introducing altered forms of one or more components of the protein complexes in order to modulate the activity of such complexes. For example, by reducing or inhibiting a complex component activity within target cells, an abnormal signal transduction event(s) leading to a condition may be decreased, inhibited, or reversed. Deletion or missense mutants of a component, that retain the ability to interact with other components of the protein complexes but cannot function in signal transduction, may be used to inhibit an abnormal, deleterious signal transduction event.
  • Expression vectors derived from viruses such as retroviruses, vaccinia virus, adenovirus, adeno-associated virus, herpes viruses, several RNA viruses, or bovine papilloma virus, may be used for delivery of nucleotide sequences (e.g., cDNA) encoding recombinant phosphatase of the invention protein into the targeted cell population (e.g., tumor cells).
  • viruses such as retroviruses, vaccinia virus, adenovirus, adeno-associated virus, herpes viruses, several RNA viruses, or bovine papilloma virus.
  • recombinant viral vectors containing coding sequences can be used to construct recombinant viral vectors containing coding sequences (Maniatis et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989; Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, N.Y., 1989).
  • recombinant nucleic acid molecules encoding protein sequences can be used as naked DNA or in a reconstituted system e.g., liposomes or other lipid systems for delivery to target cells (e.g., Felgner et al., Nature 337:387-8, 1989).
  • gene transfer can be performed by simply injecting minute amounts of DNA into the nucleus of a cell, through a process of microinjection (Capecchi, Cell 22:479-88, 1980). Once recombinant genes are introduced into a cell, they can be recognized by the cell's normal mechanisms for transcription and translation, and a gene product will be expressed. Other methods have also been attempted for introducing DNA into larger numbers of cells. These methods include: transfection, wherein DNA is precipitated with calcium phosphate and taken into cells by pinocytosis (Chen et al., Mol. Cell Biol.
  • adenovirus proteins are capable of destabilizing endosomes and enhancing the uptake of DNA into cells.
  • the admixture of adenovirus to solutions containing DNA complexes, or the binding of DNA to polylysine covalently attached to adenovirus using protein crosslinking agents substantially improves the uptake and expression of the recombinant gene (Curiel et al., Am. J. Respir. Cell. Mol. Biol., 6:247-52, 1992).
  • Gene transfer means the process of introducing a foreign nucleic acid molecule into a cell. Gene transfer is commonly performed to enable the expression of a particular product encoded by the gene.
  • the product may include a protein, polypeptide, anti-sense DNA or RNA, or enzymatically active RNA.
  • Gene transfer can be performed in cultured cells or by direct administration into animals. Generally gene transfer involves the process of nucleic acid contact with a target cell by non-specific or receptor mediated interactions, uptake of nucleic acid into the cell through the membrane or by endocytosis, and release of nucleic acid into the cytoplasm from the plasma membrane or endosome. Expression may require, in addition, movement of the nucleic acid into the nucleus of the cell and binding to appropriate nuclear factors for transcription.
  • gene therapy is a form of gene transfer and is included within the definition of gene transfer as used herein and specifically refers to gene transfer to express a therapeutic product from a cell in vivo or in vitro. Gene transfer can be performed ex vivo on cells which are then transplanted into a patient, or can be performed by direct administration of the nucleic acid or nucleic acid-protein complex into the patient.
  • a vector having nucleic acid sequences encoding a phosphatase polypeptide is provided in which the nucleic acid sequence is expressed only in specific tissue.
  • Methods of achieving tissue-specific gene expression are set forth in International Publication No. WO 93/09236, filed Nov. 3, 1992 and published May 13, 1993.
  • nucleic acid sequence contained in the vector may include additions, deletions or modifications to some or all of the sequence of the nucleic acid, as defined above.
  • Gene replacement means supplying a nucleic acid sequence which is capable of being expressed in vivo in an animal and thereby providing or augmenting the function of an endogenous gene which is missing or defective in the animal.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
  • the liposomes will be targeted to and taken up selectively by the tumor.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Suitable carriers include excipients such as, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa buffer or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be the VPD co-solvent system.
  • VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the VPD co-solvent system (VPD:D5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution.
  • This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • hydrophobic pharmaceutical compounds may be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • tyrosine or serine/threonine phosphatase modulating compounds of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • compositions suitable for use in the present invention include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the proper dosage depends on various factors such as the type of disease being treated, the particular composition being used and the size and physiological condition of the patient.
  • Therapeutically effective doses for the compounds described herein can be estimated initially from cell culture and animal models. For example, a dose can be formulated in animal models to achieve a circulating concentration range that initially takes into account the IC 50 as determined in cell culture assays. The animal model data can be used to more accurately determine useful doses in humans.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the tyrosine or serine/threonine phosphatase activity).
  • IC 50 as determined in cell culture
  • Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD 50 and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p.1).
  • Toxicity studies can also be carried out by measuring the blood cell composition.
  • toxicity studies can be carried out in a suitable animal model as follows: 1) the compound is administered to mice (an untreated control mouse should also be used); 2) blood samples are periodically obtained via the tail vein from one mouse in each treatment group; and 3) the samples are analyzed for red and white blood cell counts, blood cell composition and the percent of lymphocytes versus polymorphonuclear cells. A comparison of results for each dosing regime with the controls indicates if toxicity is present.
  • the expected daily dose of a hydrophobic pharmaceutical agent is between 1 to 500 mg/day, preferably 1 to 250 mg/day, and most preferably 1 to 50 mg/day. Drugs can be delivered less frequently provided plasma levels of the active moiety are sufficient to maintain therapeutic effectiveness.
  • Plasma levels should reflect the potency of the drug. Generally, the more potent the compound the lower the plasma levels necessary to achieve efficacy.
  • Plasma half-life and biodistribution of the drug and metabolites in the plasma, tumors and major organs can also be determined to facilitate the selection of drugs most appropriate to inhibit a disorder. Such measurements can be carried out.
  • PLC analysis can be performed on the plasma of animals treated with the drag and the location of radiolabeled compounds can be determined using detection methods such as X-ray, CAT scan and MRI.
  • Compounds that show potent inhibitory activity in the screening assays, but have poor pharmacokinetic characteristics, can be optimized by altering the chemical structure and retesting. In this regard, compounds displaying good pharmacokinetic characteristics can be used as a model.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the phosphatase modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90% inhibition of the phosphatase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, “PLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drag may not be related to plasma concentration.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the polynucleotide for human or veterinary administration.
  • Such notice for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • Suitable conditions indicated on the label may include treatment of a tumor, inhibition of angiogenesis, treatment of fibrosis, diabetes, and the like.
  • a functional derivative is meant a “chemical derivative,” “fragment,” or “variant,” of the polypeptide or nucleic acid of the invention, which terms are defined below.
  • a functional derivative retains at least a portion of the function of the protein, for example reactivity with an antibody specific for the protein, enzymatic activity or binding activity mediated through noncatalytic domains, which permits its utility in accordance with the present invention. It is well known in the art that due to the degeneracy of the genetic code numerous different nucleic acid sequences can code for the same amino acid sequence. Equally, it is also well known in the art that conservative changes in amino acid can be made to arrive at a protein or polypeptide that retains the functionality of the original. In both cases, all permutations are intended to be covered by this disclosure.
  • the present invention is intended to include any nucleic acid sequence resulting from the addition of ATG as an initiation codon at the 5′-end of the inventive nucleic acid sequence or its derivative, or from the addition of TTA, TAG or TGA as a termination codon at the 3′-end of the inventive nucleotide sequence or its derivative.
  • the nucleic acid molecule of the present invention may, as necessary, have restriction endonuclease recognition sites added to its 5′-end and/or 3′-end.
  • nucleic acid sequence affords an opportunity to promote secretion and/or processing of heterologous proteins encoded by foreign nucleic acid sequences fused thereto.
  • All variations of the nucleotide sequence of the phosphatase genes of the invention and fragments thereof permitted by the genetic code are, therefore, included in this invention.
  • a “chemical derivative” of the complex contains additional chemical moieties not normally a part of the protein.
  • Covalent modifications of the protein or peptides are included within the scope of this invention. Such modifications may be introduced into the molecule by reacting targeted amino acid residues of the peptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues, as described below.
  • Cysteinyl residues most commonly are reacted with alpha-haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives. Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p-chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, or chloro-7-nitrobenzo-2-oxa-1,3-diazole.
  • Lysinyl and amino terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect or reversing the charge of the lysinyl residues.
  • Other suitable reagents for derivatizing primary amine containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate.
  • Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pK a of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine alpha-amino group.
  • Tyrosyl residues are well-known targets of modification for introduction of spectral labels by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizol and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
  • Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
  • Derivatization with bifunctional agents is useful, for example, for cross-linking the component peptides of the protein to each other or to other proteins in a complex to a water-insoluble support matrix or to other macromolecular carriers.
  • Commonly used cross-linking agents include, for example, 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3′-dithiobis(succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane.
  • Derivatizing agents such as methyl-3-[p-azidophenyl)dithiolpropioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light.
  • reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization.
  • Such derivatized moieties may improve the stability, solubility, absorption, biological half-life, and the like.
  • the moieties may alternatively eliminate or attenuate any undesirable side effect of the protein complex and the like.
  • Moieties capable of mediating such effects are disclosed, for example, in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Co., Easton, Pa. (1990).
  • variant polypeptide which either lacks one or more amino acids or contains additional or substituted amino acids relative to the native polypeptide.
  • the variant may be derived from a naturally occurring complex component by appropriately modifying the protein DNA coding sequence to add, remove, and/or to modify codons for one or more amino acids at one or more sites of the C-terminus, N-terminus, and/or within the native sequence. It is understood that such variants having added, substituted and/or additional amino acids retain one or more characterizing portions of the native protein, as described above.
  • a functional derivative of a protein with deleted, inserted and/or substituted amino acid residues may be prepared using standard techniques well-known to those of ordinary skill in the art.
  • the modified components of the functional derivatives may be produced using site-directed mutagenesis techniques (as exemplified by Adelman et al., 1983, DNA 2:183) wherein nucleotides in the DNA coding the sequence are modified such that a modified coding sequence is modified, and thereafter expressing this recombinant DNA in a prokaryotic or eukaryotic host cell, using techniques such as those described above.
  • proteins with amino acid deletions, insertions and/or substitutions may be conveniently prepared by direct chemical synthesis, using methods well-known in the art.
  • the functional derivatives of the proteins typically exhibit the same qualitative biological activity as the native proteins.
  • the invention also provides methods for determining whether a nucleic acid sequence encodes a phosphatase, according to the invention, which contains one or more characterizing portions of the native complex.
  • examples of such retained characteristics include: catalytic activity; substrate specificity; interaction with other molecules in the intact cell; regulatory functions; or binding with an antibody specific for the native complex, or an epitope thereof.
  • the invention provides an assay analyzing one or more characteristics—in particular, the presence of a catalytic domain—of a polypeptide phosphatase encoded by a given nucleic acid molecule.
  • a suitable assay can begin by purifying and quantitating a photphase protein.
  • the protein then can be assayed, for example, by serial dilution and incubation in a buffer (e.g. ABT buffer) comprising a substrate capable of undergoing hydrolysis and optionally a reducing agent capable of increasing any catalytic activity of the polypeptide.
  • a buffer e.g. ABT buffer
  • the substrate is p-nitrophenyl phosphate (pNPP) and the reducing agent is dithiothreitol (DTT), at mM concentrations of 4 ⁇ and 1 ⁇ , respectively.
  • Incubation can be at room temperature from about 2 minutes to overnight, depending on activity.
  • NaOH which can be about 100 ul of 10 N NaOH.
  • the suspension can be centrifuged and the supernatant analyzed at an OD of 410 nM to determine whether to protein phosphatase exhibited catalytic properties.
  • Table 1 documents the name of each gene, the classification of each gene product, the positions of the open reading frames within the sequence, and the length of the corresponding peptide. From left to right the data presented is as follows: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Superfamily”, “Group”, “Family”, “NA_length”, “OUF Start”, “ORF End”, “ORF Length”, and “AA_length”. “Gene name” refers to name given the sequence encoding the phosphatase or phosphatase-like enzyme. Each gene is represented by “SGP” designation followed by an arbitrary number. The SGP name usually represents multiple overlapping sequences built into a single contiguous sequence (a “contig”).
  • Group and “Family” refer to the phosphatase classification defined by sequence homology and based on previously established phylogenetic ( The Protein Phosphatase Factsbook, Nick Tonks, Shirish Shenolikar, Harry Charbonneau, Academic Pr, 2000).
  • NA_length refers to the length in nucleotides of the corresponding nucleic acid sequence.
  • ORF start refers to the beginning nucleotide of the open reading frame.
  • ORF end refers to the last nucleotide of the open reading frame, including the stop codon.
  • ORF length refers to the length in nucleotides of the open reading frame.
  • AA length refers to the length in amino acids of the peptide encoded in the corresponding nuclei acid sequence.
  • Chrosome refers to the cytogenetic localization of the gene.
  • Information in the “SNPs” column describes the nucleic acid position and degenerate nature of candidate single nucleotide polymorphisms (SNPs.
  • “dbEST hits” lists accession numbers of entries in the public database of ESTs (dbEST, http://www.ncbi.nlm.nih.gov/dbEST/index.html) that contain at least 100 bp of 100% identity to the corresponding gene. These ESTs were identified by blastn of dbEST. “Repeats” contains information about the location of short sequences, approximately 20 bp in length, that are of low complexity and that are present in several distinct genes.
  • Table 3 lists the extent and the boundaries of the phosphatase catalytic domains.
  • the column headings are: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Domain”, “Phos_start”, “Phos_end”, “Profile_start”, “Profile_end”.
  • the contents columns “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, are as described above for Table 1.
  • “Phos_Start”, “Phos_End”, “Profile_Start” and “Profile_End” refer to data obtained using a Hidden-Markov Model to define catalytic range boundaries (http://pfam.wustl.edu/index.html).
  • Genes which have a partial catalytic domain will have a “Profile Start” of greater than 1 (indicating that the beginning of the phosphatase domain is missing, and/or a “Profile End” of less than 261 (indicating that the C-terminal end of the phosphatase domain is missing).
  • Each of the sequences encompasses a complete catalytic domain, except for SGP024, which has a partial catalytic domain represents amino acids 205 to 264 of the PTP profile.
  • “ACC#nraa_match” lists the accession number of the most similar protein in the NCBI database of non-redundant proteins. “Description” contains the name of the most similar protein in the NCBI database of non-redundant proteins. “Query start” refers to the amino acid number in the phosphatase (“Query”) at which the alignment begins. “Query end” refers to the amino acid number in the phosphatase (“Query”) at which the alignment ends. “Target start” refers to the amino acid number in the Smith Waterman hit (“Target”) from NRAA at which the alignment begins. “Target end” refers to the amino acid number in the Smith Waterman hit (“Target”) from NRAA at which the alignment ends.
  • Table 5 shows the results of a gene expression analysis of selected phosphatases presented in this application using a microarray of cDNAs derived from 499 tissues and cell lines.
  • the cDNAs were spotted on nylon and probed with labeled phosphatase genes, as described in Materials and Methods below.
  • the phosphatase probes were PCR cloned from genomic exons.
  • Table 6 “Multiple Tissue Blot”, contains results of probing a Clontech Multiple Tissue Blot with radioactively labeled probes derived from SGP002 and SGP012. The table lists the tissues on the blot and the values obtained for relative gene expression in each tissue.
  • Novel phosphatases were identified from the Celera human genomic sequence databases, and from the public Human Genome Sequencing project (http://www.ncbi.nlm.nih.gov/) using hidden Markov models (HMMRs).
  • HMMRs hidden Markov models
  • the genomic database entries were translated in six open reading frames and searched against the model using a Timelogic Decypher box with a Field programmable array (FPGA) accelerated version of HMMR2.1.
  • FPGA Field programmable array
  • the DNA sequences encoding the predicted protein sequences aligning to the MR profile were extracted from the original genomic database.
  • the nucleic acid sequences were then clustered using the Pangea Clustering tool to eliminated repetitive entries.
  • the putative protein phosphatase sequences were then sequentially run through a series of queries and filters to identity novel protein phosphatase sequences.
  • the HMMR identified sequences were searched using BLASTN and BLASTX against a nucleotide and amino acid repository containing known human protein phosphatases and all subsequent new protein phosphatase sequences as they are identified.
  • the output was parsed into a spreadsheet to facilitate elimination of known genes by manual inspection. Two models were developed, a “complete” model and a “partial” or Smith Waterman model. The partial model was used to identify sub-catalytic phosphatase domains, whereas the complete model was used to identify complete catalytic domains.
  • the selected hits were then queried using BLASTN against the public nrna and EST databases to confirm they are indeed unique. In some cases the novel genes were judged to be orthologues of previously identified rodent or vertebrate protein phosphatases.
  • Another method for defining DNA extensions from genomic sequence used iterative searches of genomic databases through the Genescan program to predict exon splicing. These predicted genes were then assessed to see if they represented “real” extensions of the partial genes based on homology to related phosphatases.
  • the N-terminal region (1-335) was derived from Genewise predictions using Celera contig 300825903, with protein homologs gi
  • Incyte sequence 339266.1 is missing exon 7 (GFSVSTAGRMHIFKPVSVQAMW).
  • 7242951 (KIAA1298) is missing exon 11 and starts near the beginning of exon 10. The lack of exon 11 causes a frameshift, and so KIAA1298 has a divergent N-terminal predicted peptide, reading exon 10 in a different frame.
  • SGP006 is identical to KIAA1298 over the C-terminal 715 amino acids of SGP006 (amino acids 335 to 1049).
  • SGP006 (SEQ ID NO: 1) is 6374 nucleotides long. The open reading frame starts at position 34 and ends at position 3183, giving an ORF length of 3150 nucleotides. The predicted protein is 1049 amino acids long. This sequence is fill length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 12q21.3-q22. Amplification of this chromosomal position has been associated with the following human diseases: Bladder carcinoma (12q21-q24, 1/16) (Knuutila, et al.).
  • ESTs for this gene in the public domain are: BE793092.1, AI651213.1, BE256978.1. This gene has repetitive sequence at the following nucleotide positions: Alu 5750-6010; 5750-5770.
  • SGP002 nucleic acid sequence was derived from Genewise algorithm run with Celera genomic DNA 70000016592596 and the protein homolog gi — 6679156. A similar Genscan prediction gave an N-terminal extension, and comparison with HGP contig gi
  • Incyte EST 1026659.2 encodes an alternative splice form missing an exon which includes part of the phosphatase domain.
  • Incyte EST 1026659.7 adds further 172 nucleotides of 5′ UTR to the gene.
  • Incyte and public ESTs show expression in many tissues, most commonly digestive system, nervous system, respiratory system, and male and female genitalia.
  • SGP002 (SEQ ID NO: 2) is 2732 nucleotides long. The open reading frame starts at position 538 and ends at position 2535, giving an ORF length of 1998 nucleotides. The predicted protein is 665 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 12p11.1-p12.1.
  • the sequence for SGP018 is predicted from Celera contig 68000017706859, using Gensca and genewise with gi — 7305011 and gi — 7705959.
  • the genewise prediction covered most of a putative phosphatase.
  • the Genscan prediction overlapped and extended the genewise predictions, and almost all of the genscan was covered by ESTs from Incyte and dbEST. In all cases, ESTs were corrected by first aligning with genomic (Colera/HGP) sequence.
  • a splice variant predicted by Genscan would replace the sequence SEFLDEALLTYR with YCHYIIFSCVFIS (changes the nt sequence ACTGTCATTACATCATTTTCTCTTGTGTTTTCATTTC to CTGAGTTCCTGGATGAGGCGCTGCTGACTTACAG.
  • EST origins Incyte sequences: 981712.1, 981712.3, 981712.2, 364575.1, 061688.1, 144608.1, 7668648H1, 7473603CB1, 7473604CB1.
  • the cDNA sequence from 544-612 is not covered by any ESTs. Accordingly, the upstrea and downstream sequences could be different genes and a start at position 613 would give a peptid a later start, at MLESAE; this would give a protein with good homology and the same N-terminal length as the closest mouse homolog, PTP13.
  • a possible alternative splice form seen by comparin incyte ESTs 061688.1 and 7668648H1 predicts a protein form which is missing the Nterminus and instead starts at the sequence MTPEK
  • SGP018 (SEQ ID NO: 4) is 4361 nucleotides long. The open reading frame starts at position 208 and ends at position 3609, giving an ORF length of 3402 nucleotides. The predicted protein is 1133 amino acids long. This sequence is fill length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene has not been mapped to a chromosomal position.
  • ss1765941; 1161 S (catotaccccaatgas) dbSNP
  • ESTs for this gene in the public domain are: BF114881. This gene has repetitive sequence at the following nucleotide positions: 1603-1627.
  • SGP003 sequence is derived from Genewise with Celera contig 173000019613519 and NCBI homolog template gi — 7705959, extended to the stop codon by genomic walk.
  • the cDNA template is built from 4 EST clones, 2 from muscle, one each from bone and parathyroid gland. Corrected a frameshift in the sequence using HGP contig gi
  • SGP003 has a 235 nucleotide open reading frame preceding the start codon, extending from nucleotide 3 to nucleotide 239, shown in capital letters below: caAGGGTTTCAGGTCGCACTGGAAAATCATTTTGCAAGCAGATGT CATAGGTCTCCTCTTAGACTGGACGGCACGCAAGGTCAGCGTCACAGATC TGACCCTAAAAATAGGCCTCTGTTGCCAGTCGGGGTGGCTGGGCGTGCGG CTGCTACATGCCCCACGGACCAGAACCTCCCGACGCGGCCAGGCCCCGGC ACACCCAGCTGCAGAAAGGAGAGAAAATCCCTTGGCTCTAAAatg
  • SGP003 (SEQ ID NO: 5) is 1262 nucleotides long. The open reading frame starts at position 240 and ends at position 902, giving an ORF length of 663 nucleotides. The predicted protein is 220 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position CHR10. This gene has repetitive sequence at the following nucleotide positions: 311-334.
  • DSP domain 1 runs from the second half of Exon1 to the end of exon3, domain 2 runs from towards the end of exon7 to almost the end of exon 9.
  • Start of exon 9 (EDG-LPT) is missing in gi
  • 2833919 have a frameshift relative to the consensus towards the end of exon 9, which replaces the sequence after NSGF with SGSSRFWTTDWGGRRGGSDLAGSQDP*.
  • This change destroys the end of the phosphatase domain, and is not similar to anything in the database. It could be due to genomic polymorphism between individuals, a repeated sequencing error, or possibly some form of gene regulation.
  • These ESTs come from testis (2, same library), prostate and cardiac, so are not a library artifact.
  • 8009838H1 has an internal deletion within exon 2 from YLG-SSA.
  • 304421CB1 is missing exons 2-6 and has a frameshift between exons 1 and 7, and is missing start of exon 9.
  • 128077.1 is missing exons 2,3,6 and the start of exon 9.
  • SGP014 (SEQ ID NO: 6) is 1917 nucleotides long. The open reading frame starts at position 31 and ends at position 1680, giving an ORF length of 1650 nucleotides. The predicted protein is 549 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 10q21.3. This chromosomal position has been associated with the following human diseases: Squamous cell carcinomas of the head and neck (10q21-q22, 2/30) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: AA723271, AW444890.1, AA435513.1.
  • SGP060 The sequence of SGP060 is derived from Genewise, using Celera contig 6514035 —1 and protein homolog NP — 057448. NCBI ESTs used to extend the sequence include BF207232, BF314818, AW953216.1.
  • SGP060 (SEQ ID NO: 7) is 636 nucleotides long. The open reading frame starts at position 1 and ends at position 636, giving an ORF length of 636 nucleotides. The predicted protein is 211 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 8p11.1-q11.1 centromeric. This chromosomal position has been associated with the following human diseases: breast carcinoma (8p11-p12, 8/53); non-small cell lung cancer (18p11.2, 2/50) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: BF207232, BF314818, AW953216.1.
  • Genscan and genewise were done on Celera contig 78000006091415, using homologs gi
  • SGP039 is derived from Celera sequence 17000030279756, and from Incyte sequences 272616.1 and 7476908CB1.
  • SGP039 (SEQ ID NO: 9) is 1083 nucleotides long. The open reading frame starts at position 1 and ends at position 1083, giving an ORF length of 1083 nucleotides. The predicted protein is 360 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Serine Phosphatase, STP, PP2C. This gene has not been mapped to a chromosomal position. ESTs for this gene in the public domain (dbEST) are: BE147139.
  • SGP040, PDP (SEQ ID NO: 10) is 1725 nucleotides long. The open reading frame starts at position 1 and ends at position 1725, giving an ORF length of 1725 nucleotides. The predicted protein is 574 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Serine Phosphatase, STP, PP2C. This gene maps to chromosomal position 8q21.3. This chromosomal position has been associated with the following human diseases: Mantle cell lymphoma (18q21-q23; 5/50) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: AV706533.1, AV705571.1, AV710801.1.
  • the sequence for SGP012 is derived from Genewise, using Celera sequences 94000002120453; 142000016367225; 142000016006753, as geneomic DNA input and NP — 031981 (murine PTP-EST) as protein homolog.
  • Incyte ESTSs that overlap this sequence include 1005303.1, and 7109651 — 3.
  • Public ESTs which overlap with the sequence include AL042532.1, AI381571, and AW872677.
  • SGP012 PTP-ESP (SEQ ID NO: 11) is 4719 nucleotides long. The open reading frame starts at position 1 and ends at position 4719, giving an ORF length of 4719 nucleotides.
  • the genomic sequence for this gene is of fairly poor quality, i.e., it has not been assembled and has apparent sequence errors. Thus the nucleic acid and protein sequences are partial, with gaps indicated by “X” s in the sequence.
  • the predicted protein is 1573 amino acids long. This sequence contains the catalytic domain. It is classified as (superfamily/group/family): Tyrosine Phosphatase, RPTm, PTPd. This gene has not been mapped to chromosomal position.
  • ESTs for this gene in the public domain are: AL042532.1, AI381571, AW872677. This gene has repetitive sequence at the following nucleotide positions: 1305-1324.
  • SGP024 is derived from Genewise using Celera DNA sequence 142000016226692 as geneomic source and NP — 002830.1 (human PTP delta) as protein homolog.
  • SGP024 (SEQ ID NO: 12) is 354 nucleotides long. The open reading frame starts at position 1 and ends at position 357, giving an ORF length of 357 nucleotides. The predicted protein is 118 amino acids long. This sequence is a partail catalytic domain. It is classified as (superfamily/group/family): Tyrosine Phosphatase, Receptor PTP, PTPdelta sub-family.
  • SGP006, KIAA1298 (SEQ ID NO: 1) encodes SEQ ID NO: 13, a protein that is 1049 amino acids long. It is classified as an MKP.
  • the phosphatase domain in this protein matches the hidden Markov profile for a MKP/DSP phosphatase from profile position 1 to profile position 173 (full length catalytic domain).
  • the position of the catalytic region within the encoded protein is from amino acid 308 to amino acid 446.
  • the results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results.
  • the C-terminus of SGP006 (amino acid positions 322 to 1049) is 100% identical to KIAA1298 protein [ Homo sapiens ].
  • SGP002 (SEQ ID NO: 2) encodes SEQ ID NO: 14, a protein that is 665 amino acids long. It is classified as an MKP.
  • the phosphatase domain in this protein matches the hidden Markov MKP/DSP phosphatase domain from profile position 1 to profile position 173 (full length catalytic domain).
  • the position of the catalytic region within the encoded protein is from amino acid 158 to amino acid 297.
  • SGP002 is regulated in response to the cellular redox environment (Nandi et al., Int J Biochem Cell Biol April 2000; 32(4):465-73; Rhodanese as a thioredoxin oxidase ).
  • SGP001 (SEQ ID NO: 3) encodes SEQ ID NO: 15, a protein that is 498 amino acids long. It is classified as an MKP.
  • the phosphatase domain in this protein matches the hidden Markov profile for a MKP/DSP phosphatase domain from profile position 1 to profile position 173.
  • the position of the catalytic region within the encoded protein is from amino acid 307 to amino acid 441.
  • SGP014 (SEQ ID NO: 6) encodes SEQ ID NO: 18, a protein that is 549 amino acids long, with two phosphatase domains. Both domains in this protein match the hidden Markov profile for an MKP/DSP phosphatase profile from position 1 to profile position 173 (fall length). Both DSP domains are similar, with best hits to gi
  • SGP060 (SEQ ID NO: 7) encodes SEQ ID NO: 19, a protein that is 211 amino acids long. It is classified as an MKP.
  • the phosphatase domain in this protein matches the hidden Markov profile for MKP/DSP phosphatase from profile position 1 to profile position 173 (full length).
  • the position of the catalytic region within the encoded protein is from amino acid 61 to amino acid 204.
  • SGP039 (SEQ ID NO: 9) encodes SEQ ID NO: 21, a protein that is 360 amino acids long. It is classified as: PP2C,.
  • the phosphatase domain in this protein matches the hidden Markov profile from profile position 1 to profile position 301 (Full length catalytic).
  • the position of the catalytic region within the encoded protein is from amino acid 91 to amino acid 344.
  • SGP040 PDP (SEQ ID NO: 10) encodes SEQ ID NO: 22, a protein that is 574 amino acids long. It is classified as: PP2C.
  • the phosphatase domain in this protein matches the hidden Markov profile from position 1 to position 301.
  • the position of the catalytic region within the encoded protein is from amino acid 209 to amino acid 497.
  • SGP012 PTP-ESP (SEQ ID NO: 11) encodes SEQ ID NO: 23, a protein that is 1573 amino acids long. It is classified as: PTP, delta phosphatase-like. The phosphatase domain in this protein matches the hidden Markov profile for a PTP phosphatase, from profile position 1 to profile position 264 (full length catalytic). The position of the catalytic region within the encoded protein is from amino acid 1010 to 1259.
  • SGP024 (SEQ ID NO: 12) encodes SEQ ID NO: 24, a protein that is 118 amino acids long. It is classified as a PTP, related to PTP delta.
  • the phosphatase domain in this protein matches the hidden Markov profile for a PTP from profile position 205 to profile position 264 (this is a partial catalytic domain, representing the C-terminal region).
  • the position of the catalytic region within the encoded protein is from amino acid 3 to amino acid 58.
  • cDNA libraries derived from a variety of sources were immobilized onto nylon membranes and probed with 32P-labeled cDNA fragments derived from the gene(s) of interest.
  • the sources of RNA are listed in Table 3. They are: 1) Biochain Institute (Hayward, Calif.; http://www.biochain.com/main — 3.html): 2) Clontech (Palo Alto, Calif., http://www.clontech.com/); 3) mammalian cell lines used by the National Cancer Institute (NCI) Developmental Therapeutics Program (http://dtp.nci.nih.gov/; can be ordered from ATCC: http://www.atcc.org/catalogs.html): 4) PathAssociates http://www.saic.com/company/subsidiaries/pai.html; San Diego, Calif.).
  • the protocols for preparing cDNA arrays are detailed below.
  • Several cell lines were treated with compounds to evaluate their effects on gene expression. There were eight treatments: 1) control, 2) low sereum, 3) 200 uM mimosine, 4) 3 mM HU, 5) 2 uM AUR2 inhibitor,) 10 uM cisplatin, 7) 400 ng/ml nocodozole-24 hours, and 8) 400 ng/ml nocodozole-48 hours.
  • the treated cell lines are listed by cell line name followed by a number from 1 to 8.
  • cDNA libraries derived from over 450 tissue or cell line sources were immobilized onto nylon membranes and probed with 32P-labeled cDNA fragments derived from the gene(s) of interest.
  • total RNA or mRNA was used as template in a reverse transcription reaction to generate single-stranded cDNAs (ss cDNA) that were tagged with specific sequences at each end.
  • the synthesized cDNAs contain specific sequence tags at both the 5′ and the 3′ end.
  • the 5′ and the 3′ ends are tagged with the same sequence (CDS and SMII) it is referred to as “symmetric”.
  • the 5′ end is tagged with a different sequence than the 3′ end (CDS and ML2G) is referred to as “asymmetric”.
  • a double-stranded “cDNA library” is then generated by PCR amplification using the 3′PCR and ML2 primers (3′ PCR: AAGCAGTGGTAACAACGCAGAGT and ML2: AAGTGGCAACAGAGATAACGCGT) that anneal to the added sequence tags.
  • the amplified “cDNA libraries” were manually arrayed onto nylon membranes with a 384 pin replicator.
  • the DNA was denatured by alkali treatment, neutralized and cross-linked by UV light.
  • the arrays were pre-hybridized with Express Hyb (Clontech) and hybridized with 32 p labeled probes generated by random hexamer priming of cDNA fragments corresponding to the genes of interest. After washing, the blots were exposed to phosphorimaging cassettes and the intensity of the signal was quantified.
  • the amount of the DNA on the arrays was also quantified by treating non-denatured or denatured arrays with Syber Green I or Syber Green II respectively (1:100,000 in 50 mM Tris, pH8.0) for 2 minutes. After washing with 50 mM Tris, pH8.0, the fluorescent emission was detected with a phosphorimager (Molecular Dynamics) and quantified. The amount of the arrayed DNA was used to normalize the hybridization signal and the corrected values are tabulated in Table 5.
  • tissue array data for the 3 phosphatases were standardized for statistical analysis across the different tissue types using range standardization. Standardization converts measurements to a common scale. We used range standardization, which subtracts the smallest value of each variable from each value and divides by its range. The new scale starts at 0 and ends at 1.0. The following statistical procedures were implemented on the standardized data: generation of descriptive statistics, graphical visualization, hierarchical and k-means cluster analysis (at 10, 7, and 5 clusters), and comparison of groups using analysis of variance (ANOVA). When tissue-specific data were present for both normal and tumor samples, the two groups were directly compared for fold differences.
  • Cluster 1 (singleton).
  • NORMAL GROUP heart sample (tissue).
  • Cluster 2 (singleton).
  • NORMAL GROUP spinal cord (tissue).
  • NORMAL GROUP only: colon (stomach tissue), colon (small intestinal tissue), mammary epithelial cells (cell line), spleen (heme tissue), lymph node theme tissue), fetal lung (tissue), fetal brain (neural tissue), and prostate (tissue).
  • Cluster 1 (singleton).
  • NORMAL GROUP adrenal gland (tissue).
  • NORMAL GROUP thymus (heme tissue).
  • TUMOR GROUP lung carcinoma (cell line), and a neuro sample (tissue).
  • TUMOR GROUP only: lung (tissue), neuroblastoma (tissue), and colon carcinoma (cell line).
  • TUMOR GROUP MG (tissue), smc (cell line), glioblastoma (cell line), lung large cell carcinoma (cell line), END (tissue), primary renal cell carcinoma (cell line), and lung (tissue); and NORMAL GROUP: brain (tissue).
  • Cluster 1 (singleton).
  • TUMOR GROUP neuro (tissue).
  • TUMOR GROUP only: HNS (tissue), renal adenocarcinoma (cell line), and ovary (cell line).
  • TUMOR GROUP only: malignant melanoma, metastasis to lung (cell line), colon (cell line, treated with 3 mM HEW), ovary (cell line, treated with 10 uM cisplatin), neuro (cell line, treated with 10 uM cisplatin), and PML peripheral blood, promyelocytic leukemia (cell line).
  • TUMOR GROUP colon (cell line, treated with 10 uM cisplatin), breast (cell line), endothelial cells (cell line, treated with HeLa25X DEF-MES for hypoxia, 4 hours), unknown sample (unknown), cervical (cell line, treated with 400 ng/ml noco-48 hours), kidney (tissue), lung (tissue), lung (tissue), endothelial cells (cell line), and lung (tissue); and NORMAL GROUP: HUVEC (cell line, treated with 10 mn PDGF stimulation).
  • TUMOR GROUP kidney carcinoma (cell line), lung (tissue), neuro (cell line, treated with 10 uM cisplatin), lung (tissue), bone (cell line), breast (cell line), lung (tissue), lung (cell line, treated with 3 mM HU), neuro (cell line, treated with 400 ng/ml noco-24 hours), endothelial cells (cell line, treated with HeLa25X DEF-MES for hypoxia, 0 hours), ovary (cell line, treated with 2 uM AUR2 inhibitor), breast (cell line, treated with normal/10% FBS), breast (cell line, treated with 2 uM AUR2 inhibitor), breast (cell line, treated with 200 uM mimosine), bone (cell line, treated with low serim/0.1% EBS), colon (cell line, treated with 10 uM cisplatin), cervical (cell line, treated with low serim/0.1% FBS), endothelial cells (cell line), kidney (tissue), pancreas
  • MTE (Multiple Tissue Expression) blots were obtained from Clontech Laboratories, Inc (see table 6). These blots contained 84 arrayed cDNA samples derived from normal human tissue and human cell lines, and controls. The expression blots were prehybridized with ExpressHyb hybridization solution (Clontech Laboratories) containing 0.1 mg/ml denatured salmon sperm DNA at a temperature of 65° C. for two hours. Radioactive DNA probes were prepared using the Random Priming DNA labeling kit (Roche). Purified DNA fragments (100 ng) were labeled with 250 uCi of 32P-labeled dCTP for 45 minutes using the kit protocol.
  • Unincorporated nucleotide was removed through the use of a spin column (ProbeQuant G50 micro columns, Amersham Pharmacia, Inc.). After denaturation by boiling for three minutes, the probe was introduced into the prehybridization solution, and the blot was hybridized at 65° C. for 20 hours. The blot was subsequently washed four times for 15 minutes each at 65° C. in a solution containing 15 mM NaCl, 1.5 mM Na 3 Citrate, 0.1% sodiumn lauryl sulfate (SDS) and exposed to the phosphoimager screen for quantitation.
  • SDS sodiumn lauryl sulfate
  • SGP012 (SEQ ID NO: 11, encoding SEQ ID NO: 23) is expressed at the highest levels in the following tissues: testis; cerebellum, right; colon, descending; cerebellum left; lymph node; Burkitt's lymphoma; Daudi; and mammary gland. This pattern of expression suggests that SGP012 may play a role in diseases of the central nervous system (cerebellum exprssion), in immune system disease (the lymph node, Burkitt's lymphoma, and Daudi are all immune system tissues), or breast cancer (from expression in mammary tissue).
  • SGP002 (SEQ ID NO: 2, encoding SEQ ID NO: 14) is expressed at the highest levels in the following tissues: adrenal gland; placenta; prostate; salivary gland; mary gland; pituitary gland. Expression in the prostate and breast may indicate a role for this phosphatase in cancer of these tissues. Expression in the adrenal gland may indicate a role in metabolic processes controled by that gland, such as stress response.
  • OMIM maintains a searchable catalog of cytogenetic map locations organized by disease.
  • Medline http://www.ncbi.nlm.nih.gov/PubMed/medline.html.
  • References for association of the mapped sites with chromosomal abnormalities found in human cancer can be found in: Knuutila, et al., Am J Pathol, 1998, 152:1107-1123. The results are discussed in the Section on Nucleic Acids above.
  • SNPs were also identified by comparing several databases of expressed genes (dbEST, NRNA) and genomic sequence (i.e., NRNA) for single basepair mismatches. The results are shown in Table 2, in the column labeled “SNPs”. These are candidate SNPs—their actual frequency in the human population was not determined.
  • C Cytidine
  • R G or A
  • puRine Y C or T
  • pYrimidine K G or T
  • Keto W A or T
  • S C or G
  • M A or C
  • aMino B C, G or T (i.e., not A)
  • D A, G or T (i.e., not C)
  • H A, C or T (i.e., not G)
  • V A, C or G (i.e., not T)
  • N A, C, G or T
  • aNy X A, C, G or T
  • the dbSNP accession number is rs881179. This SNP occurs in the 3′ untranslated region.
  • RNAs are isolated using the Guanidine Salts/Phenol extraction protocol of Chomczynski and Sacchi (P. Chomczynski and N. Sacchi, Anal. Biochem. 162, 156 (1987)) from primary human tumors, normal and tumor cell lines, normal human tissues, and sorted human hematopoietic cells. These RNAs are used to generate single-stranded cDNA using the Superscript Preamplification System (GIBCO BRL, Gaithersburg, Md.; Gerard, G F et al. (1989), FOCUS 11, 66) under conditions recommended by the manufacturer. A typical reaction uses 10 ⁇ g total RNA with 1.5 ⁇ g oligo(dT) 12-18 in a reaction volume of 60 ⁇ L. The product is treated with RNaseH and diluted to 100 ⁇ L with H 2 O. For subsequent PCR amplification, 1-4 ⁇ L of this sscDNA is used in each reaction.
  • PCR reactions are performed using degenerate primers applied to multiple single-stranded cDNAs.
  • the primers are added at a final concentration of 5 ⁇ M each to a mixture containing 10 mM TrisHCl, pH 8.3, 50 mM KCl, 1.5 mM MgCl 2 , 200 ⁇ M each deoxynucleoside triphosphate, 0.001% gelatin, 1.5 U AmpliTaq DNA Polymerase (Perkin-Elmer/Cetus), and 1-4 ⁇ L cDNA.
  • the cycling conditions are 94° C. for 30 s, 50° C. for 1 min, and 72° C. for 1 min 45 s for 35 cycles.
  • PCR fragments migrating between 300-350 bp are isolated from 2% agarose gels using the GeneClean Kit (Bio101), and T-A cloned into the pCRII vector (Invitrogen Corp. U.S.A.) according to the manufacturer's protocol.
  • Colonies are selected for mini plasmid DNA-preparations using Qiagen columns and the plasmid DNA is sequenced using a cycle sequencing dye-terminator kit with AmpliTaq DNA Polymerase, FS (ABI, Foster City, Calif.). Sequencing reaction products are run on an ABI Prism 377 DNA Sequencer, and analyzed using the BLAST alignment algorithm (Altschul, S. F. et al., J. Mol. Biol. 215: 403-10).
  • Probes are 32 P-labeled by random priming and used at 2 ⁇ 10 6 cpm/mL following standard techniques for library screening. Pre-hybridization (3 h) and hybridization (overnight) are conducted at 42° C. in 5 ⁇ SSC, 5 ⁇ Denhart's solution, 2.5% dextran sulfate, 50 mM Na 2 PO 4 /NaHPO 4 , pH 7.0, 50% formamide with 100 mg/mL denatured salmon sperm DNA. Stringent washes are performed at 65° C. in 0.1 ⁇ SSC and 0.1% SDS. DNA sequencing is carried out on both strands using a cycle sequencing dye-terminator kit with AmpliTaq DNA Polymerase, FS (ABI, Foster City, Calif.). Sequencing reaction products are run on an ABI Prism 377 DNA Sequencer.
  • Expression constructs are generated for some of the human cDNAs including: a) full-length clones in a pCDNA expression vector; b) a GST-fusion construct containing the catalytic domain of the novel phosphatase fused to the C-terminal end of a GST expression cassette; and c) a full-length clone containing a Cys to Ser (C to S) mutation at the predicted catalytic site within the phosphatase domain, inserted in the pCDNA vector.
  • Proteins in SDS PAGE are transferred to immobilon membrane.
  • the washing buffer is PBST (standard phosphate-buffered saline pH 7.4+0.1% Triton X-100).
  • Blocking and antibody incubation buffer is PBST +5% milk.
  • Antibody dilutions varied from 1:1000 to 1:2000.
  • the pcDNA expression plasmids (10 ⁇ g DNA/100 mm plate) containing the STE20-related phosphatase constructs are introduced into 293 cells with lipofectamine (Gibco BRL). After 72 hours, the cells are harvested in 0.5 mL solubilization buffer (20 mM HEPES, pH 7.35, 150 mM NaCl, 10% glycerol, 1% Triton X-100, 1.5 mM MgCl 2 , 1 mM EGTA, 2 mM phenylmethylsulfonyl fluoride, 1 ⁇ g/mL aprotinin).
  • solubilization buffer (20 mM HEPES, pH 7.35, 150 mM NaCl, 10% glycerol, 1% Triton X-100, 1.5 mM MgCl 2 , 1 mM EGTA, 2 mM phenylmethylsulfonyl fluoride, 1 ⁇ g/mL
  • phosphatase inhibitors (10 mM NaBPO 4 , pH 7.25, 150 mM NaCl, 1% Triton X-100, 0.5% deoxycholate, 0.1% SDS, 0.2% sodium azide, 1 mM NaF, 1 mM EGTA, 4 mM sodium orthovanadate, 1% aprotinin, 5 ⁇ g/mL leupeptin).
  • Cell debris is removed by centrifugation (12000 ⁇ g, 15 min, 4° C.) and the lysate is precleared by two successive incubations with 50 ⁇ L of a 1:1 slurry of protein A sepharose for 1 hour each.
  • One-half mL of the cleared supernatant is reacted with 10 ⁇ L of protein A purified phosphatase-specific antisera (generated from the GST fusion protein or antipeptide antisera) plus 50 ⁇ L of a 1:1 slurry of protein A-sepharose for 2 hr at 4° C.
  • the beads are then washed 2 times in PBSTDS, and 2 times in HNTG (20 mM HEPES, pH 7.5/150 mM NaCl, 0.1% Triton X-100, 10% glycerol).
  • the immunopurified phosphatases on sepharose beads are resuspended in 20 ⁇ L HNTG plus 30 mM MgCl 2 , 10 mM MnCl 2 , and 20 ⁇ Ci [ ⁇ 32 P]ATP (3000 Ci/mmol).
  • the phosphatase reactions are run for 30 min at room temperature, and stopped by addition of HNTG supplemented with 50 mM EDTA.
  • the samples are washed 6 times in HNTG, boiled 5 min in SDS sample buffer and analyzed by 6% SDS-PAGE followed by autoradiography. Phosphoamino acid analysis is performed by standard 2D methods on 32 P-labeled bands excised from the SDS-PAGE gel.
  • Nylon membranes are purchased from Boehringer Mannheim. Denaturing solution contains 0.4 M NaOH and 0.6 M NaCl. Neutralization solution contains 0.5 M Tris-HCL, pH 7.5 and 1.5 M NaCl. Hybridization solution contains 50% formamide, 6 ⁇ SSPE, 2.5 ⁇ Denhardt's solution, 0.2 mg/nl denatured salmon DNA, 0.1 mg/mL yeast tRNA, and 0.2% sodium dodecyl sulfate. Restriction enzymes are purchased from Boebringer Mannheim. Radiolabeled probes are prepared using the Prime-it II kit by Stratagene. The beta-actin DNA fragment used for a probe template is purchased from Clontech.
  • Genomic DNA is isolated from a variety of tumor cell lines (such as MCF-7, MDA-MB-231, Calu-6, A549, HCT-15, HT-29, Colo 205, LS-180, DLD-1, HCT-116, PC3, CAPAN-2, MIA-PaCa-2, PANC-1, AsPc-1, BxPC-3, OVCAR-3, SKOV3, SW 626 and PA-1, and from two normal cell lines.
  • tumor cell lines such as MCF-7, MDA-MB-231, Calu-6, A549, HCT-15, HT-29, Colo 205, LS-180, DLD-1, HCT-116, PC3, CAPAN-2, MIA-PaCa-2, PANC-1, AsPc-1, BxPC-3, OVCAR-3, SKOV3, SW 626 and PA-1, and from two normal cell lines.
  • a 10 ⁇ g aliquot of each genomic DNA sample is digested with EcoR I restriction enzyme and a separate 10 ⁇ g sample is digested with Hind III restriction enzyme.
  • the restriction-digested DNA samples are loaded onto a 0.7% agarose gel and, following electrophoretic separation, the DNA is capillary-transferred to a nylon membrane by standard methods (Sambrook, J. et al (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory).
  • bound phage is eluted in 100 ⁇ L of 1% SDS and plated on agarose plates to obtain single plaques.
  • the present invention relates to compounds demonstrating the ability to modulate protein enzymes related to cellular signal transduction; preferably, protein phosphatases; and most preferably, protein tyrosine phosphatases.
  • the assays described below are employed to select those compounds demonstrating the optimal degree of the desired activity.
  • the phrase “optimal degree of desired activity” refers to the highest therapeutic index, defined above, against a protein enzyme which mediates cellular signal transduction and which is related to a particular disorder so as to provide an animal or a human patient, suffering from such disorder with a therapeutically effective amount of a compound of this invention at the lowest possible dosage.
  • phosphatases such assays involve exposing target cells in culture to the compounds and (a) biochemically analyzing cell lysates to assess the level and/or identity of phosphorylated proteins; or (b) scoring phenotypic or functional changes in treated cells as compared to control cells that were not exposed to the test substance.
  • the cells are exposed to the compound of the invention and compared to positive controls which are exposed only to the natural ligand, and to negative controls which are not exposed to either the compound or the natural ligand.
  • the assay may be carried out in the absence of the ligand.
  • the cells are exposed to the compound of the invention in the presence of the natural ligand and compared to controls which are not exposed to the compound of the invention.
  • the assays described below may be used as a primary screen to evaluate the ability of the compounds of this invention to inhibit phosphatase activity of the compounds of the invention.
  • the assays may also be used to assess the relative potency of a compound by testing a range of concentrations, in a range from 100 ⁇ M to 1 pM, for example, and computing the concentration at which the amount of phosphorylation or signal transduction is reduced or increased by 50% (IC50) compared to controls.
  • the protein components separated by SDS-PAGE are transferred to a nitrocellulose membrane, the presence of pTyr is detected using an antiphosphotyrosine (anti-pTyr) antibody.
  • anti-pTyr antiphosphotyrosine
  • the substrate of interest be first isolated by incubating the cell lysate with a substrate-specific anchoring antibody bound to a solid support, and thereafter, washing away non-bound cellular components, and assessing the presence or absence of pTyr on the solid support by an anti-pTyr antibody.
  • This preferred method can readily be performed in a microtiter plate format by an automated robotic system, allowing for testing of large numbers of samples within a reasonably short time frame.
  • the anti-pTyr antibody can be detected by labeling it with a radioactive substance which facilitates its detection by autoradiography.
  • the anti-pTyr antibody can be conjugated with an enzyme, such as horseradish peroxidase, and detected by subsequent addition of an appropriate substrate for the enzyme, the choice of which would be clear to one skilled in the art.
  • a further alternative involves detecting the anti-pTyr antibody by reacting with a second antibody which recognizes the anti-pTyr antibody, this second antibody being labeled with either a radioactive substance or an enzyme as previously described. Any other methods for the detection of an antibody known in the art may be used.
  • the above methods may also be used in a cell-free system wherein cell lysate containing the signal-transducing substrate molecule and phosphatase is mixed with a compound of the invention and a kinase.
  • the substrate is phosphorylated by initiating the kinase reaction by the addition of adenosine triphosphate (ATP).
  • ATP adenosine triphosphate
  • the reaction mixture may be analyzed by the SDS-PAGE technique or it may be added to a substrate-specific anchoring antibody bound to a solid support, and a detection procedure as described above is performed on the separated or captured substrate to assess the presence or absence of pTyr. The results are compared to those obtained with reaction mixtures to which the compound is not added.
  • the cell-free system does not require the natural ligand or knowledge of its identity.
  • Posner et al. U.S. Pat. No. 5,155,031 describes the use of insulin receptor as a substrate and rat adipocytes as target cells to demonstrate the ability of pervanadate to inhibit PTP activity.
  • vanadate has been shown to activate an insulin-sensitive facilitated transport system for glucose and glucose analogs in rat adipocytes (Dubyak et al., 1980, J. Biol. Chem., 256:5306-5312).
  • the activity of the compounds of the invention may be assessed by measuring the increase in the rate of transport of glucose analog such as 2-deoxy- 3 H-glucose in rat adipocytes that have been exposed to the compounds.
  • Vanadate also mimics the effect of insulin on glucose oxidation in rat adipocytes (Shechter et al., 1980, Nature, 284:556-558).
  • the compounds of this invention may be tested for stimulation of glucose oxidation by measuring the conversion of 14 C-glucose to 14 CO 2 .
  • the effect of sodium orthovanadate on erythropoietin-mediated cell proliferation has been measured by cell cycle analysis based on DNA content as estimated by incorporation of tritiated thymidine during DNA synthesis (Spivak et al., 1992, Exp. Hematol., 20:500-504).
  • the activity of the compounds of this invention toward phosphatases that play a role in cell proliferation may be assessed by cell cycle analysis.
  • This assay may be used to test the ability of the compounds of the invention to inhibit dephosphorylation of phosphotyrosine (pTyr) residues on insulin receptor (IR).
  • IR insulin receptor
  • substrate molecules such as platelet derived growth factor receptor
  • the activities of the compounds of this invention toward different protein tyrosine enzymes may be assessed.
  • an endogenous kinase activity is active at low level even in the absence of insulin binding. Thus, no insulin is needed to stimulate phosphorylation of IR.
  • cell lysates can be prepared and added to microtiter plates coated with anti-insulin receptor antibody.
  • the level of phosphorylation of the captured insulin receptor is detected using an anti-pTyr antibody and an enzyme-linked secondary antibody.
  • the catalytic assays described herein are performed in a 96-well format.
  • the general procedure begins with the determination of PTP optimal pH using a three-component buffer system that minimizes ionic strength variations across a wide range of buffer pH.
  • the Michaelis-Menten constant, or Km is determined for each specific substrate-PTP system. This Km value is subsequently used as the substrate reaction concentration for compound screening.
  • the test PTP is exposed to varying concentrations of compound for fifteen minutes and allowed to react with substrate for ten minutes. The results are plotted as percent inhibition versus compound concentration and the IC50 interpolated from the plot.
  • nucleic acid sequences encode the claimed peptides and proteins of the invention.
  • all four nucleic acid sequences GCT, GCC, GCA, and GCG encode the amino acid alanine. Therefore, if for an amino acid there exists an average of three codons, a polypeptide of 100 amino acids in length will, on average, be encoded by 3100, or 5 ⁇ 1047, nucleic acid sequences.
  • a nucleic acid sequence can be modified to form a second nucleic acid sequence, encoding the same polypeptide as encoded by the first nucleic acid sequences, using routine procedures and without undue experimentation.
  • nucleic acids that encode the claimed peptides and proteins are also fully described herein, as if all were written out in full taking into account the codon usage, especially that preferred in humans.
  • changes in the amino acid sequences of polypeptides, or in the corresponding nucleic acid sequence encoding such polypeptide may be designed or selected to take place in an area of the sequence where the significant activity of the polypeptide remains unchanged. For example, an amino acid change may take place within a ⁇ -turn, away from the active site of the polypeptide.
  • changes such as deletions (e.g. removal of a segment of the polypeptide, or in the corresponding nucleic acid sequence encoding such polypeptide, which does not affect the active site) and additions (e.g.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to phosphatase polypeptides, nucleotide sequences encoding the phosphatase polypeptides, as well as various products and methods useful for the diagnosis and treatment of various phosphatase-related diseases and conditions. Through the use of a bioinformatics strategy, mammalian members of the MAP kinase hosphatase PTP's and STP's have been identified and their protein structure predicted.

Description

  • The present invention claims priority on provisional application serial Nos. 60/173,255, 60/178,078, 60/179,301, 60/175,766, (and the provisional application serial no. represented by Sugen docket no. “[0001] Cel 16”), all of which are hereby incorporated by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to phosphatase polypeptides, nucleotide sequences encoding the phosphatase polypeptides, as well as various products and methods useful for the diagnosis and treatment of various phosphatase-related diseases and conditions. [0002]
  • BACKGROUND OF THE INVENTION
  • The following description of the background of the invention is provided to aid in understanding the invention, but is not admitted to be or to describe prior art to the invention. [0003]
  • Cellular signal transduction is a fundamental mechanism whereby external stimuli that regulate diverse cellular processes are relayed to the interior of cells. One of the key biochemical mechanisms of signal transduction involves the reversible phosphorylation of proteins by protein kinases, which enables regulation of the activity of mature proteins by altering their structure and function. The best characterized protein kinases in eukaryotes phosphorylate proteins on the alcohol moiety of serine, threonine and tyrosine residues. These kinases largely fall into two groups: those specific for phosphorylating serines and threonines, and those specific for phosphorylating tyrosines. [0004]
  • The phosphorylation state of a given substrate is also regulated by the protein phosphatases, a class of proteins responsible for removal of the phosphate group added to a given substrate by a protein kinase. The protein phosphatases can also be classified as being specific for either serine/threonine or tyrosine. Some members of this family are able to dephosphorylate only tyrosine, and are known as the “protein tyrosine phosphatases” (“CTP”); while others are able to dephosphorylate tyrosine as well as serine and threonine, and are named, “dual-specificity phosphatases” (“DSP”); and a third family dephosphorylates only serine or threonine (“STP”)—as disclosed by Fauman et al., [0005] Trends Biochem. Sci. November 1996;21(11):413-7; and Martell et al., Mol. Cells. Feb. 28, 1998;8(1): 2-11. These proteins share a 250-300 amino acid domain that comprises the common catalytic core structure. Related phosphatases are clustered into distinct subfamilies of tyrosine phosphatases, dual-specificity phosphatases, and myotubularin-like phosphatases (Fauman et al., supra; and Martell et al., supra).
  • Phosphatases possess a variety of non-catalytic domains that are believed to interact with upstream regulators. Examples include proline-rich domains for interaction with SH3-containing proteins, or specific domains for interaction with Rac, Rho, and Rab small G-proteins. These interactions may provide a mechanism for cross-talk between distinct biochemical pathways in response to external stimuli such as the activation of a variety of cell surface receptors, including tyrosine kinases, cytokine receptors, TNF receptor, Fas, T cell receptors, CD28, or CD40. [0006]
  • Phosphatases have been implicated as regulating a variety of cellular responses, including response to growth factors, cytokines and hormones, oxidative-, UV-, or irradiation-related stress pathways, inflammatory signals (e.g. TNFα), apoptotic stimuli (e.g. Fas), T and B cell costimulation, the control of cytoskeletal architecture, and cellular transformation (see THE PROTEIN PHOSPHATASE FACTBOOK, Tonks et al., Academic Press, 2000). [0007]
  • A need, therefore, exists to identify additional phosphatases whose inappropriate activity may lead to cancer or other disorders so that appropriate treatments for those disorders might also be identified. [0008]
  • SUMMARY OF THE INVENTION
  • The following abbreviations are use to describe characeristics of the phosphatases according to the invention: [0009]
  • DsPTP Dual specificity protein phosphatase [0010]
  • DUS Dual specificity phosphatase [0011]
  • MKP MAP Kinase phosphatase [0012]
  • MTM Myotubular myopathy (myotubularin) phosphatase [0013]
  • PTP Protein Tyrosine Phosphatase [0014]
  • PTEN Phosphatase and tensin homolog [0015]
  • Through the use of a “motif extraction” bioinformatics script, the named inventors have identified certain mammalian members of the phosphatase family, which are disclosed herein. The invention provides a partial or complete sequence of 12 phosphatases, as well as the classification, predicted or deduced protein structure, and a strategy for elucidating the biologic and therapeutic relevance of these proteins. These novel proteins include: eight (8) MAP kinase phosphatase enzymes (“MKPs”), which are members of the DSP family; two (2) phosphatases from the STP family; and two (2) phosphatases from the PTP family. The classification of novel proteins as belonging to established families has proven highly accurate, not only in predicting motifs present in the remaining non-catalytic portion of each protein, but also in the regulation, substrates, and signaling pathways fo these proteins. [0016]
  • One aspect of the invention features an identified, isolated, enriched, or purified nucleic acid molecule encoding a phosphatase polypeptide, having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. [0017]
  • By “isolated” in reference to nucleic acid is meant a polymer of 10 (preferably 21, more preferably 39, most preferably 75) or more nucleotides conjugated to each other, including DNA and RNA that is isolated from a natural source or that is synthesized as the sense or complementary antisense strand. In certain embodiments of the invention, longer nucleic acids are preferred, for example those of 300, 600, 900, 1200, 1500, or more nucleotides and/or those having at least 50%, 60%, 75%, 90%, 95% or 99% identity to a sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12. [0018]
  • The isolated nucleic acid of the present invention is unique in the sense that it is not found in a pure or separated state in nature. Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular (i.e., chromosomal) environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment. The term does not imply that the sequence is the only nucleotide chain present, but that it is essentially free (about 90-95% pure at least) of non-nucleotide material naturally associated with it, and thus is distinguished from isolated chromosomes. [0019]
  • By the use of the term “enriched” in reference to nucleic acid is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction (2- to 5-fold) of the total DNA or RNA present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of other DNA or RNA present, or by a preferential increase in the amount of the specific DNA or RNA sequence, or by a combination of the two. However, it should be noted that enriched does not imply that there are no other DNA or RNA sequences present, just that the relative amount of the sequence of interest has been significantly increased. The term “significant” is used to indicate that the level of increase is useful to the person making such an increase, and generally means an increase relative to other nucleic acids of about at least 2-fold, more preferably at least 5- to 10-fold or even more. The term also does not imply that there is no DNA or RNA from other sources. The DNA from other sources may, for example, comprise DNA from a yeast or bacterial genome, or a cloning vector such as pUC19. This term distinguishes from naturally occurring events, such as viral infection, or tumor-type growths, in which the level of one mRNA may be naturally increased relative to other species of mRNA. That is, the term is meant to cover only those situations in which a person has intervened to elevate the proportion of the desired nucleic acid. [0020]
  • It is also advantageous for some purposes that a nucleotide sequence be in purified form. The term “purified” in reference to nucleic acid does not require absolute purity (such as a homogeneous preparation). Instead, it represents an indication that the sequence is relatively more pure than in the natural environment (compared to the natural level this level should be at least 2- to 5-fold greater, e.g., in terms of mg/mL). Individual clones isolated from a cDNA library may be purified to electrophoretic homogeneity. The claimed DNA molecules obtained from these clones could be obtained directly from total DNA or from total RNA. The cDNA clones are not naturally occurring, but rather are preferably obtained via manipulation of a partially purified naturally occurring substance (messenger RNA). The construction of a cDNA library from mRNA involves the creation of a synthetic substance (cDNA) and pure individual cDNA clones can be isolated from the synthetic library by clonal selection of the cells carrying the cDNA library. Thus, the process which includes the construction of a cDNA library from mRNA and isolation of distinct cDNA clones yields an approximately 10[0021] 6-fold purification of the native message. Thus, purification of at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated.
  • By a “phosphatase polypeptide” is meant 32 (preferably 40, more preferably 45, most preferably 55) or more contiguous amino acids in a polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. In certain aspects, polypeptides of 100, 200, 300, 400, 450, 500, 550, 600, 700, 800, 900 or more amino acids are preferred. The phosphatase polypeptide can be encoded by a full-length nucleic acid sequence or any portion of the full-length nucleic acid sequence, so long as a functional activity of the polypeptide is retained. It is well known in the art that due to the degeneracy of the genetic code numerous different nucleic acid sequences can code for the same amino acid sequence. Equally, it is also well known in the art that conservative changes in amino acid can be made to arrive at a protein or polypeptide which retains the functionality of the original. Such substitutions may include the replacement of an amino acid by a residue having similar physicochemical properties, such as substituting one aliphatic residue (Ile, Val, Leu or Ala) for another, or substitution between basic residues Lys and Arg, acidic residues Glu and Asp, amide residues Gln and Asn, hydroxyl residues Ser and Tyr, or aromatic residues Phe and Tyr. Further information regarding making amino acid exchanges which have only slight, if any, effects on the overall protein can be found in Bowie et al., [0022] Science, 1990, 247:1306-1310, which is incorporated herein by reference in its entirety including any figures, tables, or drawings. In all cases, all permutations are intended to be covered by this disclosure.
  • The amino acid sequence of the phosphatase peptide of the invention will be substantially similar to a sequence having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or the corresponding full-length amino acid sequence, or fragments thereof. [0023]
  • A sequence that is substantially similar to a sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24 will preferably have at least 90% identity (more preferably at least 95% and most preferably 99-100%) to the sequence. [0024]
  • By “identity” is meant a property of sequences that measures their similarity or relationship. Identity is measured by dividing the number of identical residues by the total number of residues and gaps and multiplying the product by 100. “Gaps” are spaces in an alignment that are the result of additions or deletions of amino acids. Thus, two copies of exactly the same sequence have 100% identity, but sequences that are less highly conserved, and have deletions, additions, or replacements, may have a lower degree of identity. Those skilled in the art will recognize that several computer programs are available for determining sequence identity using standard parameters, for example Gapped BLAST or PSI-BLAST (Altschul, et al. (1997) Nucleic Acids Res. 25:3389-3402), BLAST (Altschul, et al. (1990) J. Mol. Biol. 215:403-410), and Smith-Waterman (Smith, et al. (1981) J. Mol. Biol. 147:195-197). Preferably, the default settings of these programs will be employed, but those skilled in the art recognize whether these settings need to be changed and know how to make the changes. [0025]
  • “Similarity” is measured by dividing the number of identical residues plus the number of conservatively substituted residues (see Bowie, et al. [0026] Science, 1999 247:1306-1310, which is incorporated herein by reference in its entirety, including any drawings, figures, or tables) by the total number of residues and gaps and multiplying the product by 100.
  • In preferred embodiments, the invention features isolated, enriched, or purified nucleic acid molecules encoding a phosphatase polypeptide comprising a nucleotide sequence that: (a) encodes a polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; (b) is the complement of the nucleotide sequence of (a); (c) hybridizes under highly stringent conditions to the nucleotide molecule of (a) and encodes a naturally occurring phosphatase polypeptide; (d) encodes a polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, except that it lacks one or more, but not all, of the domains selected from the group consisting of an N-terminal domain, a catalytic domain, a C-terminal catalytic domain, a C-terminal domain, a coiled-coil structure region, a proline-rich region, a spacer region, and a C-terminal tail; and (e) is the complement of the nucleotide sequence of (d). [0027]
  • The term “complement” refers to two nucleotides that can form multiple favorable interactions with one another. For example, adenine is complementary to thymine as they can form two hydrogen bonds. Similarly, guanine and cytosine are complementary since they can form three hydrogen bonds. A nucleotide sequence is the complement of another nucleotide sequence if all of the nucleotides of the first sequence are complementary to all of the nucleotides of the second sequence. [0028]
  • Various low or high stringency hybridization conditions may be used depending upon the specificity and selectivity desired. These conditions are well known to those skilled in the art. Under stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having more than 1 or 2 mismatches out of 20 contiguous nucleotides, more preferably, such conditions prevent hybridization of nucleic acids having more than 1 or 2 mismatches out of 50 contiguous nucleotides, most preferably, such conditions prevent hybridization of nucleic acids having more than 1 or 2 mismatches out of 100 contiguous nucleotides. In some instances, the conditions may prevent hybridization of nucleic acids having more than 5 mismatches in the full-length sequence. [0029]
  • By stringent hybridization assay conditions is meant hybridization assay conditions at least as stringent as the following: hybridization in 50% formamide, 5×SSC, 50 mM NaH[0030] 2PO4, pH 6.8, 0.5% SDS, 0.1 mg/mL sonicated salmon sperm DNA, and 5× Denhardt's solution at 42° C. overnight; washing with 2×SSC, 0.1% SDS at 45° C.; and washing with 0.2×SSC, 0.1% SDS at 45° C. Under some of the most stringent hybridization assay conditions, the second wash can be done with 0.1×SSC at a temperature up to 70° C. (Berger et al. (1987) Guide to Molecular Cloning Techniques pg 421, hereby incorporated by reference herein in its entirety including any figures, tables, or drawings.). However, other applications may require the use of conditions falling between these sets of conditions. Methods of determining the conditions required to achieve desired hybridizations are well known to those with ordinary skill in the art, and are based on several factors, including but not limited to, the sequences to be hybridized and the samples to be tested. Washing conditions of lower stringency frequently utilize a lower temperature during the washing steps, such as 65° C., 60° C., 55° C., 50° C., or 42° C.
  • The term “domain” refers to a region of a polypeptide which serves a particular function. For instance, N-terminal or C-terminal domains of signal transduction proteins can serve functions including, but not limited to, binding molecules that localize the signal transduction molecule to different regions of the cell or binding other signaling molecules directly responsible for propagating a particular cellular signal. Some domains can be expressed separately from the rest of the protein and function by themselves, while others must remain part of the intact protein to retain function. The latter are termed functional regions of proteins and also relate to domains. [0031]
  • The term “N-terminal domain” refers to the extracatalytic region located between the initiator methionine and the catalytic domain of the protein phosphatase. The N-terminal domain can be identified following a Smith-Waterman alignment of the protein sequence against the non-redundant protein database to define the N-terminal boundary of the catalytic domain. Depending on its length, the N-terminal domain may or may not play a regulatory role in phosphatase function. The term “catalytic domain” refers to a region of the protein phosphatase that is typically 25-300 amino acids long and is responsible for carrying out the phosphate transfer reaction from a high-energy phosphate donor molecule such as ATP or GTP to itself (autophosphorylation) or to other proteins (exogenous phosphorylation). The catalytic domain of protein phosphatases is made up of 12 subdomains that contain highly conserved amino acid residues, and are responsible for proper polypeptide folding and for catalysis. The catalytic domain can be identified following a Smith-Waterman alignment of the protein sequence against the non-redundant protein database. [0032]
  • The term “catalytic activity”, as used herein, defines the rate at which a phosphatase catalytic domain phosphorylates a substrate. Catalytic activity can be measured, for example, by determining the amount of a substrate converted to a phosphorylated product as a function of time. Catalytic activity can be measured by methods of the invention by holding time constant and determining the concentration of a phosphorylated substrate after a fixed period of time. Phosphorylation of a substrate occurs at the active site of a protein phosphatase. The active site is normally a cavity in which the substrate binds to the protein phosphatase and is phosphorylated. [0033]
  • The term “substrate” as used herein refers to a molecule phosphorylated by a phosphatase of the invention. Phosphatases phosphorylate substrates on serine/threonine or tyrosine amino acids. The molecule may be another protein or a polypeptide. [0034]
  • The term “C-terminal domain” refers to the region located between the catalytic domain or the last (located closest to the C-terminus) functional domain and the carboxy-terminal amino acid residue of the protein phosphatase. By “functional” domain is meant any region of the polypeptide that may play a regulatory or catalytic role as predicted from amino acid sequence homology to other proteins or by the presence of amino acid sequences that may give rise to specific structural conformations (e.g. N-terminal domain). The C-terminal domain can be identified by using a Smith-Waterman alignment of the protein sequence against the non-redundant protein database to define the C-terminal boundary of the catalytic domain or of any functional C-terminal extracatalytic domain. Depending on its length and amino acid composition, the C-terminal domain may or may not play a regulatory role in phosphatase function. For the some of the phosphatases of the instant invention, the C-terminal domain may also comprise the catalytic domain (above). [0035]
  • The term “C-terminal tail” as used herein, refers to a C-terminal domain of a protein phosphatase, that by homology extends or protrudes past the C-terminal amino acid of its closest homolog. C-terminal tails can be identified by using a Smith-Waterman sequence alignment of the protein sequence against the non-redundant protein database, or by means of a multiple sequence alignment of homologous sequences using the DNAStar program Megalign. Depending on its length, a C-terminal tail may or may not play a regulatory role in phosphatase function. [0036]
  • The term “coiled-coil structure region” as used herein, refers to a polypeptide sequence that has a high probability of adopting a coiled-coil structure as predicted by computer algorithms such as COILS (Lupas, A. (1996) [0037] Meth. Enzymology 266:513-525). Coiled-coils are formed by two or three amphipathic α-helices in parallel. Coiled-coils can bind to coiled-coil domains of other polypeptides resulting in homo- or heterodimiers (Lupas, A. (1991) Science 252:1162-1164).
  • The term “proline-rich region” as used herein, refers to a region of a protein phosphatase whose proline content over a given amino acid length is higher than the average content of this amino acid found in proteins (i.e., >10%). Proline-rich regions are easily discemable by visual inspection of amino acid sequences and quantitated by standard computer sequence analysis programs such as the DNAStar program EditSeq. Proline-rich regions have been demonstrated to participate in regulatory protein-protein interactions. [0038]
  • The term “spacer region” as used herein, refers to a region of the protein phosphatase located between predicted functional domains. The spacer region has no detectable homology to any amino acid sequence in the database, and can be identified by using a Smith-Waterman alignment of the protein sequence against the non-redundant protein database to define the C- and N-terminal boundaries of the flanking functional domains. Spacer regions may or may not play a fundamental role in protein phosphatase function. [0039]
  • The term “insert” as used herein refers to a portion of a protein phosphatase that is absent from a close homolog. Inserts may or may not by the product alternative splicing of exons. Inserts can be identified by using a Smith-Waterman sequence alignment of the protein sequence against the non-redundant protein database, or by means of a multiple sequence alignment of homologous sequences using the DNAStar program Megalign. Inserts may play a functional role by presenting a new interface for protein-protein interactions, or by interfering with such interactions. [0040]
  • The term “signal transduction pathway” refers to the molecules that propagate an extracellular signal through the cell membrane to become an intracellular signal. This signal can then stimulate a cellular response. The polypeptide molecules involved in signal transduction processes are typically receptor and non-receptor protein tyrosine phosphatases, receptor and non-receptor protein phosphatases, polypeptides containing [0041] SRC homology 2 and 3 domains, phosphotyrosine binding proteins (SRC homology 2 (SH2) and phosphotyrosine binding (PTB and PH) domain containing proteins), proline-rich binding proteins (SH3 domain containing proteins), GTPases, phosphodiesterases, phospholipases, prolyl isomerases, proteases, Ca2+ binding proteins, cAMP binding proteins, guanyl cyclases, adenylyl cyclases, NO generating proteins, nucleotide exchange factors, and transcription factors.
  • In other preferred embodiments, the invention features isolated, enriched, or purified nucleic acid molecules encoding phosphatase polypeptides, further comprising a vector or promoter effective to initiate transcription in a host cell. The invention also features recombinant nucleic acid, preferably in a cell or an organism. The recombinant nucleic acid may contain a sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or a functional derivative thereof, and a vector or a promoter effective to initiate transcription in a host cell. The recombinant nucleic acid can alternatively contain a transcriptional initiation region functional in a cell, a sequence complementary to an RNA sequence encoding a phosphatase polypeptide and a transcriptional termination region functional in a cell. Specific vectors and host cell combinations are discussed herein. [0042]
  • The term “vector” relates to a single or double-stranded circular nucleic acid molecule that can be transfected into cells and replicated within or independently of a cell genome. A circular double-stranded nucleic acid molecule can be cut and thereby linearized upon treatment with restriction enzymes. An assortment of nucleic acid vectors, restriction enzymes, and the knowledge of the nucleotide sequences cut by restriction enzymes are readily available to those skilled in the art. A nucleic acid molecule encoding a phosphatase can be inserted into a vector by cutting the vector with restriction enzymes and ligating the two pieces together. [0043]
  • The term “transfecting” defines a number of methods to insert a nucleic acid vector or other nucleic acid molecules into a cellular organism. These methods involve a variety of techniques, such as treating the cells with high concentrations of salt, an electric field, detergent, or DMSO to render the outer membrane or wall of the cells permeable to nucleic acid molecules of interest or use of various viral transduction strategies. [0044]
  • The term “promoter” as used herein, refers to nucleic acid sequence needed for gene sequence expression. Promoter regions vary from organism to organism, but are well known to persons skilled in the art for different organisms. For example, in prokaryotes, the promoter region contains both the promoter (which directs the initiation of RNA transcription) as well as the DNA sequences which, when transcribed into RNA, will signal synthesis initiation. Such regions will normally include those 5′-non-coding sequences involved with initiation of transcription and translation, such as the TATA box, capping sequence, CAAT sequence, and the like. [0045]
  • In preferred embodiments, the isolated nucleic acid comprises, consists essentially of, or consists of a nucleic acid sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, which encodes an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, a functional derivative thereof, or at least 35, 40, 45, 50, 60, 75, 100, 200, or 300 contiguous amino acids selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. The nucleic acid may be isolated from a natural source by cDNA cloning or by subtractive hybridization. The natural source may be mammalian, preferably human, blood, semen, or tissue, and the nucleic acid may be synthesized by the triester method or by using an automated DNA synthesizer. [0046]
  • The term “mammal” refers preferably to such organisms as mice, rats, rabbits, guinea pigs, sheep, and goats, more preferably to cats, dogs, monkeys, and apes, and most preferably to humans. [0047]
  • In yet other preferred embodiments, the nucleic acid is a conserved or unique region, for example those useful for: the design of hybridization probes to facilitate identification and cloning of additional polypeptides, the design of PCR probes to facilitate cloning of additional polypeptides, obtaining antibodies to polypeptide regions, and designing antisense oligonucleotides. [0048]
  • By “conserved nucleic acid regions”, are meant regions present on two or more nucleic acids encoding a phosphatase polypeptide, to which a particular nucleic acid sequence can hybridize under lower stringency conditions. Examples of lower stringency conditions suitable for screening for nucleic acid encoding phosphatase polypeptides are provided in Wahl et al. [0049] Meth. Enzym. 152:399-407 (1987) and in Wahl et al. Meth. Enzym. 152:415-423 (1987), which are hereby incorporated by reference herein in its entirety, including any drawings, figures, or tables. Preferably, conserved regions differ by no more than 5 out of 20 nucleotides, even more preferably 2 out of 20 nucleotides or most preferably 1 out of 20 nucleotides.
  • By “unique nucleic acid region” is meant a sequence present in a nucleic acid coding for a phosphatase polypeptide that is not present in a sequence coding for any other naturally occurring polypeptide. Such regions preferably encode 32 (preferably 40, more preferably 45, most preferably 55) or more contiguous amino acids set forth in a full-length amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. In particular, a unique nucleic acid region is preferably of mammalian origin. [0050]
  • Another aspect of the invention features a nucleic acid probe for the detection of nucleic acid encoding a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24 in a sample. The nucleic acid probe contains a nucleotide base sequence that will hybridize to the sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or a functional derivative thereof. [0051]
  • In preferred embodiments, the nucleic acid probe hybridizes to nucleic acid encoding at least 12, 32, 75, 90, 105, 120, 150, 200, 250, 300 or 350 contiguous amino acids of a full-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or a functional derivative thereof. [0052]
  • Methods for using the probes include detecting the presence or amount of phosphatase RNA in a sample by contacting the sample with a nucleic acid probe under conditions such that hybridization occurs and detecting the presence or amount of the probe bound to phosphatase RNA. The nucleic acid duplex formed between the probe and a nucleic acid sequence coding for a phosphatase polypeptide may be used in the identification of the sequence of the nucleic acid detected (Nelson et al., in Nonisotopic DNA Probe Techniques, Academic Press, San Diego, Kricka, ed., p. 275, 1992, hereby incorporated by reference herein in its entirety, including any drawings, figures, or tables). Kits for performing such methods may be constructed to include a container means having disposed therein a nucleic acid probe. [0053]
  • In another aspect, the invention describes a recombinant cell or tissue comprising a nucleic acid molecule encoding a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. In such cells, the nucleic acid may be under the control of the genomic regulatory elements, or may be under the control of exogenous regulatory elements including an exogenous promoter. By “exogenous” it is meant a promoter that is not normally coupled in vivo transcriptionally to the coding sequence for the phosphatase polypeptides. [0054]
  • The polypeptide is preferably a fragment of the protein encoded by a full-length amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. By “fragment,” is meant an amino acid sequence present in a phosphatase polypeptide. Preferably, such a sequence comprises at least 32, 45, 50, 60, 100, 200, or 300 contiguous amino acids of a fill-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ D NO: 24. [0055]
  • In another aspect, the invention features an isolated, enriched, or purified phosphatase polypeptide having the amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ED NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. [0056]
  • By “isolated” in reference to a polypeptide is meant a polymer of 6 (preferably 12, more preferably 18, most preferably 25, 32, 40, or 50) or more amino acids conjugated to each other, including polypeptides that are isolated from a natural source or that are synthesized. In certain aspects, longer polypeptides are preferred, such as those with 100, 200, 300, 400, 450, 500, 550, 600, 700, 800, 900 or more contiguous amino acids of a full-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. [0057]
  • The isolated polypeptides of the present invention are unique in the sense that they are not found in a pure or separated state in nature. Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment. The term does not imply that the sequence is the only amino acid chain present, but that it is essentially free (about 90-95% pure at least) of non-amino acid-based material naturally associated with it. [0058]
  • By the use of the term “enriched” in reference to a polypeptide is meant that the specific amino acid sequence constitutes a significantly higher fraction (2- to 5-fold) of the total amino acid sequences present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of other amino acid sequences present, or by a preferential increase in the amount of the specific amino acid sequence of interest, or by a combination of the two. However, it should be noted that enriched does not imply that there are no other amino acid sequences present, just that the relative amount of the sequence of interest has been significantly increased. The term significant here is used to indicate that the level of increase is useful to the person making such an increase, and generally means an increase relative to other amino acid sequences of about at least 2-fold, more preferably at least 5- to 10-fold or even more. The term also does not imply that there is no amino acid sequence from other sources. The other source of amino acid sequences may, for example, comprise amino acid sequence encoded by a yeast or bacterial genome, or a cloning vector such as pUC19. The term is meant to cover only those situations in which man has intervened to increase the proportion of the desired amino acid sequence. [0059]
  • It is also advantageous for some purposes that an amino acid sequence be in purified form. The term “purified” in reference to a polypeptide does not require absolute purity (such as a homogeneous preparation); instead, it represents an indication that the sequence is relatively purer than in the natural environment. Compared to the natural level this level should be at least 2- to 5-fold greater (e.g., in terms of mg/mL). Purification of at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated. The substance is preferably free of contamination at a functionally significant level, for example 90%, 95%, or 99% pure. [0060]
  • In preferred embodiments, the phosphatase polypeptide is a fragment of the protein encoded by a full-length amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ED NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO:20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. Preferably, the phosphatase polypeptide contains at least 32, 45, 50, 60, 100, 200, or 300 contiguous amino acids of a full-length sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or a functional derivative thereof. [0061]
  • In preferred embodiments, the phosphatase polypeptide comprises an amino acid sequence having (a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; and (b) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, except that it lacks one or more of the domains selected from the group consisting of a C-terminal catalytic domain, an N-terminal domain, a catalytic domain, a C-terminal domain, a coiled-coil structure region, a proline-rich region, a spacer region, and a C-terminal tail. [0062]
  • The polypeptide can be isolated from a natural source by methods well-known in the art. The natural source may be mammalian, preferably human, blood, semen, or tissue, and the polypeptide may be synthesized using an automated polypeptide synthesizer. [0063]
  • In some embodiments the invention includes a recombinant phosphatase polypeptide having (a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. By “recombinant phosphatase polypeptide” is meant a polypeptide produced by recombinant DNA techniques such that it is distinct from a naturally occurring polypeptide either in its location (e.g., present in a different cell or tissue than found in nature), purity or structure. Generally, such a recombinant polypeptide will be present in a cell in an amount different from that normally observed in nature. [0064]
  • The polypeptides to be expressed in host cells may also be fusion proteins which include regions from heterologous proteins. Such regions may be included to allow, e.g., secretion, improved stability, or facilitated purification of the polypeptide. For example, a sequence encoding an appropriate signal peptide can be incorporated into expression vectors. A DNA sequence for a signal peptide (secretory leader) may be fused in-frame to the polynucleotide sequence so that the polypeptide is translated as a fusion protein comprising the signal peptide. A signal peptide that is functional in the intended host cell promotes extracellular secretion of the polypeptide. Preferably, the signal sequence will be cleaved from the polypeptide upon secretion of the polypeptide from the cell. Thus, preferred fusion proteins can be produced in which the N-terminus of a phosphatase polypeptide is fused to a carrier peptide. [0065]
  • In one embodiment, the polypeptide comprises a fusion protein which includes a heterologous region used to facilitate purification of the polypeptide. Many of the available peptides used for such a function allow selective binding of the fusion protein to a binding partner. A preferred binding partner includes one or more of the IgG binding domains of protein A are easily purified to homogeneity by affinity chromatography on, for example, IgG-coupled Sepharose. Alternatively, many vectors have the advantage of carrying a stretch of histidine residues that can be expressed at the N-terminal or C-terminal end of the target protein, and thus the protein of interest can be recovered by metal chelation chromatography. A nucleotide sequence encoding a recognition site for a proteolytic enzyme such as enterophosphatase, factor X procollagenase or thrombin may immediately precede the sequence for a phosphatase polypeptide to permit cleavage of the fusion protein to obtain the mature phosphatase polypeptide. Additional examples of fusion-protein binding partners include, but are not limited to, the yeast I-factor, the honeybee melatin leader in sf9 insect cells, 6-His tag, thioredoxin tag, hemaglutinin tag, GST tag, and OmpA signal sequence tag. As will be understood by one of skill in the art, the binding partner which recognizes and binds to the peptide may be any ion, molecule or compound including metal ions (e.g., metal affinity columns), antibodies, or fragments thereof, and any protein or peptide which binds the peptide, such as the FLAG tag. [0066]
  • In another aspect, the invention features an antibody (e.g., a monoclonal or polyclonal antibody) having specific binding affinity to a phosphatase polypeptide or a phosphatase polypeptide domain or fragment where the polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. By “specific binding affinity” is meant that the antibody binds to the target phosphatase polypeptide with greater affinity than it binds to other polypeptides under specified conditions. Antibodies or antibody fragments are polypeptides that contain regions that can bind other polypeptides. The term “specific binding affinity” describes an antibody that binds to a phosphatase polypeptide with greater affinity than it binds to other polypeptides under specified conditions. Antibodies can be used to identify an endogenous source of phosphatase polypeptides, to monitor cell cycle regulation, and for immuno-localization of phosphatase polypeptides within the cell. [0067]
  • The term “polyclonal” refers to antibodies that are heterogenous populations of antibody molecules derived from the sera of animals immunized with an antigen or an antigenic functional derivative thereof. For the production of polyclonal antibodies, various host animals may be immunized by injection with the antigen. Various adjuvants may be used to increase the immunological response, depending on the host species. [0068]
  • “Monoclonal antibodies” are substantially homogenous populations of antibodies to a particular antigen. They may be obtained by any technique which provides for the production of antibody molecules by continuous cell lines in culture. Monoclonal antibodies may be obtained by methods known to those skilled in the art (Kohler et al., [0069] Nature 256:495-497, 1975, and U.S. Pat. No. 4,376,110, both of which are hereby incorporated by reference herein in their entirety including any figures, tables, or drawings).
  • The term “antibody fragment” refers to a portion of an antibody, often the hypervariable region and portions of the surrounding heavy and light chains, that displays specific binding affinity for a particular molecule. A hypervariable region is a portion of an antibody that physically binds to the polypeptide target. [0070]
  • Antibodies or antibody fragments having specific binding affinity to a phosphatase polypeptide of the invention may be used in methods for detecting the presence and/or amount of phosphatase polypeptide in a sample by probing the sample with the antibody under conditions suitable for phosphatase-antibody immunocomplex formation and detecting the presence and/or amount of the antibody conjugated to the phosphatase polypeptide. Diagnostic kits for performing such methods may be constructed to include antibodies or antibody fragments specific for the phosphatase as well as a conjugate of a binding partner of the antibodies or the antibodies themselves. [0071]
  • An antibody or antibody fragment with specific binding affinity to a phosphatase polypeptide of the invention can be isolated, enriched, or purified from a prokaryotic or eukaryotic organism. Routine methods known to those skilled in the art enable production of antibodies or antibody fragments, in both prokaryotic and eukaryotic organisms. Purification, enrichment, and isolation of antibodies, which are polypeptide molecules, are described above. [0072]
  • Antibodies having specific binding affinity to a phosphatase polypeptide of the invention may be used in methods for detecting the presence and/or amount of phosphatase polypeptide in a sample by contacting the sample with the antibody under conditions such that an immunocomplex forms and detecting the presence and/or amount of the antibody conjugated to the phosphatase polypeptide. [0073]
  • Diagnostic kits for performing such methods may be constructed to include a first container containing the antibody and a second container having a conjugate of a binding partner of the antibody and a label, such as, for example, a radioisotope. [0074]
  • The diagnostic kit may also include notification of an FDA approved use and instructions therefor. [0075]
  • In another aspect, the invention features a hybridoma which produces an antibody having specific binding affinity to a phosphatase polypeptide or a phosphatase polypeptide domain, where the polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. By “hybridoma” is meant an immortalized cell line that is capable of secreting an antibody, for example an antibody to a phosphatase of the invention. In preferred embodiments, the antibody to the phosphatase comprises a sequence of amino acids that is able to specifically bind a phosphatase polypeptide of the invention. [0076]
  • In another aspect, the present invention is also directed to kits comprising antibodies that bind to a polypeptide encoded by any of the nucleic acid molecules described above, and a negative control antibody. [0077]
  • The term “negative control antibody” refers to an antibody derived from similar source as the antibody having specific binding affinity, but where it displays no binding affinity to a polypeptide of the invention. [0078]
  • In another aspect, the invention features a phosphatase polypeptide binding agent able to bind to a phosphatase polypeptide selected from the group having (a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. The binding agent is preferably a purified antibody that recognizes an epitope present on a phosphatase polypeptide of the invention. Other binding agents include molecules that bind to phosphatase polypeptides and analogous molecules that bind to a phosphatase polypeptide. Such binding agents may be identified by using assays that measure phosphatase binding partner activity. [0079]
  • The invention also features a method for screening for human cells containing a phosphatase polypeptide of the invention or an equivalent sequence. The method involves identifying the novel polypeptide in human cells using techniques that are routine and standard in the art, such as those described herein for identifying the phosphatases of the invention (e.g., cloning, Southern or Northern blot analysis, in situ hybridization, PCR amplification, etc.). [0080]
  • In another aspect, the invention features methods for identifying a substance that modulates phosphatase activity comprising the steps of: (a) contacting a phosphatase polypeptide selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24 with a test substance; (b) measuring the activity of said polypeptide; and (c) determining whether said substance modulates the activity of said polypeptide. [0081]
  • The term “modulates” refers to the ability of a compound to alter the function of a phosphatase of the invention. A modulator preferably activates or inhibits the activity of a phosphatase of the invention depending on the concentration of the compound exposed to the phosphatase. [0082]
  • The term “modulates” also refers to altering the function of phosphatases of the invention by increasing or decreasing the probability that a complex forms between the phosphatase and a natural binding partner. A modulator preferably increases the probability that such a complex forms between the phosphatase and the natural binding partner, more preferably increases or decreases the probability that a complex forms between the phosphatase and the natural binding partner depending on the concentration of the compound exposed to the phosphatase, and most preferably decreases the probability that a complex forms between the phosphatase and the natural binding partner. [0083]
  • The term “activates” refers to increasing the cellular activity of the phosphatase. The term inhibit refers to decreasing the cellular activity of the phosphatase. Phosphatase activity is preferably the interaction with a natural binding partner. [0084]
  • The term “complex” refers to an assembly of at least two molecules bound to one another. Signal transduction complexes often contain at least two protein molecules bound to one another. [0085]
  • The term “natural binding partner” refers to polypeptides, lipids, small molecules, or nucleic acids that bind to phosphatases in cells. A change in the interaction between a phosphatase and a natural binding partner can manifest itself as an increased or decreased probability that the interaction forms, or an increased or decreased concentration of phosphatase/natural binding partner complex. [0086]
  • The term “contacting” as used herein refers to mixing a solution comprising the test compound with a liquid medium bathing the cells of the methods. The solution comprising the compound may also comprise another component, such as dimethyl sulfoxide (DMSO), which facilitates the uptake of the test compound or compounds into the cells of the methods. The solution comprising the test compound may be added to the medium bathing the cells by utilizing a delivery apparatus, such as a pipette-based device or syringe-based device. [0087]
  • In another aspect, the invention features methods for identifying a substance that modulates phosphatase activity in a cell comprising the steps of: (a) expressing a phosphatase polypeptide in a cell, wherein said polypeptide is selected from the group having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; (b) adding a test substance to said cell; and (c) monitoring a change in cell phenotype or the interaction between said polypeptide and a natural binding partner. [0088]
  • The term “expressing” as used herein refers to the production of phosphatases of the invention from a nucleic acid vector containing phosphatase genes within a cell. The nucleic acid vector is transfected into cells using well known techniques in the art as described herein. [0089]
  • Another aspect of the instant invention is directed to methods of identifying compounds that bind to phosphatase polypeptides of the present invention, comprising contacting the phosphatase polypeptides with a compound, and determining whether the compound binds the phosphatase polypeptides. Binding can be determined by binding assays which are well known to the skilled artisan, including, but not limited to, gel-shift assays, Western blots, radiolabeled competition assay, phage-based expression cloning, co-fractionation by chromatography, co-precipitation, cross linking, interaction trap/two-hybrid analysis, southwestern analysis, ELISA, and the like, which are described in, for example, [0090] Current Protocols in Molecular Biology, 1999, John Wiley & Sons, N.Y., which is incorporated herein by reference in its entirety. The compounds to be screened include, but are not limited to, compounds of extracellular, intracellular, biological or chemical origin.
  • The methods of the invention also embrace compounds that are attached to a label, such as a radiolabel (e.g., [0091] 125I, 35S, 32P, 33P, 3H), a fluorescence label, a chemiluminescent label, an enzymic label and an immunogenic label. The phosphatase polypeptides employed in such a test may either be free in solution, attached to a solid support, borne on a cell surface, located intracellularly or associated with a portion of a cell. One skilled in the art can, for example, measure the formation of complexes between a phosphatase polypeptide and the compound being tested. Alternatively, one skilled in the art can examine the diminution in complex formation between a phosphatase polypeptide and its substrate caused by the compound being tested.
  • Other assays can be used to examine enzymatic activity including, but not limited to, photometric, radiometric, HPLC, electrochemical, and the like, which are described in, for example, [0092] Enzyme Assays: A Practical Approach, eds. R. Eisenthal and M. J. Danson, 1992, Oxford University Press, which is incorporated herein by reference in its entirety.
  • Another aspect of the present invention is directed to methods of identifying compounds which modulate (i.e., increase or decrease) activity of a phosphatase polypeptide comprising contacting the phosphatase polypeptide with a compound, and determining whether the compound modifies activity of the phosphatase polypeptide. These compounds are also referred to as “modulators of protein phosphatases.” The activity in the presence of the test compound is measured to the activity in the absence of the test compound. Where the activity of a sample containing the test compound is higher than the activity in a sample lacking the test compound, the compound will have increased the activity. Similarly, where the activity of a sample containing the test compound is lower than the activity in the sample lacking the test compound, the compound will have inhibited the activity. [0093]
  • The present invention is particularly useful for screening compounds by using a phosphatase polypeptide in any of a variety of drug screening techniques. The compounds to be screened include, but are not limited to, extracellular, intracellular, biological or chemical origin. The phosphatase polypeptide employed in such a test may be in any form, preferably, free in solution, attached to a solid support, borne on a cell surface or located intracellularly. One skilled in the art can, for example, measure the formation of complexes between a phosphatase polypeptide and the compound being tested. Alternatively, one skilled in the art can examine the diminution in complex formation between a phosphatase polypeptide and its substrate caused by the compound being tested. [0094]
  • The activity of phosphatase polypeptides of the invention can be determined by, for example, examining the ability to bind or be activated by chemically synthesised peptide ligands. Alternatively, the activity of the phosphatase polypeptides can be assayed by examining their ability to bind metal ions such as calcium, hormones, chemokines, neuropeptides, neurotransmitters, nucleotides, lipids, odorants, and photons. Thus, modulators of the phosphatase polypeptide's activity may alter a phosphatase function, such as a binding property of a phosphatase or an activity such as signal transduction or membrane localization. [0095]
  • In various embodiments of the method, the assay may take the form of a yeast growth assay, an Aequorin assay, a Luciferase assay, a mitogenesis assay, a MAP Phosphatase activity assay, as well as other binding or function-based assays of phosphatase activity that are generally known in the art. In several of these embodiments, the invention includes any of the receptor and non-receptor protein tyrosine phosphatases, receptor and non-receptor protein phosphatases, polypeptides containing [0096] SRC homology 2 and 3 domains, phosphotyrosine binding proteins (SRC homology 2 (SH2) and phosphotyrosine binding (PTB and PH) domain containing proteins), proline-rich binding proteins (SH3 domain containing proteins), GTPases, phosphodiesterases, phospholipases, prolyl isomerases, proteases, Ca2+ binding proteins, cAMP binding proteins, guanyl cyclases, adenylyl cyclases, NO generating proteins, nucleotide exchange factors, and transcription factors. Biological activities of phosphatases according to the invention include, but are not limited to, the binding of a natural or a synthetic ligand, as well as any one of the functional activities of phosphatases known in the art. Non-limiting examples of phosphatase activities include transmembrane signaling of various forms, which may involve phosphatase binding interactions and/or the exertion of an influence over signal transduction.
  • The modulators of the invention exhibit a variety of chemical structures, which can be generally grouped into mimetics of natural phosphatase ligands, and peptide and non-peptide allosteric effectors of phosphatases. The invention does not restrict the sources for suitable modulators, which may be obtained from natural sources such as plant, animal or mineral extracts, or non-natural sources such as small molecule libraries, including the products of combinatorial chemical approaches to library construction, and peptide libraries. [0097]
  • The use of cDNAs encoding phosphatases in drug discovery programs is well-known; assays capable of testing thousands of unknown compounds per day in high-throughput screens (HTSs) are thoroughly documented. The literature is replete with examples of the use of radiolabelled ligands in HTS binding assays for drug discovery (see Williams, [0098] Medicinal Research Reviews, 1991, 11, 147-184.; Sweetnam, et al., J. Natural Products, 1993, 56, 441-455 for review). Recombinant receptors are preferred for binding assay HTS because they allow for better specificity (higher relative purity), provide the ability to generate large amounts of receptor material, and can be used in a broad variety of formats (see Hodgson, Bio/Technology, 1992, 10, 973-980; each of which is incorporated herein by reference in its entirety).
  • A variety of heterologous systems is available for functional expression of recombinant receptors that are well known to those skilled in the art. Such systems include bacteria (Strosberg, et al., [0099] Trends in Pharmacological Sciences, 1992, 13, 95-98), yeast bausch, Trends in Biotechnology, 1997, 15, 487-494), several kinds of insect cells (Vanden Broeck, Int. Rev. Cytology, 1996, 164, 189-268), amphibian cells (Jayawickreme et al., Current Opinion in Biotechnology, 1997, 8, 629-634) and several mammalian cell lines (CHO, HEK293, COS, etc.; see Gerhardt, et al., Eur. J. Pharmacology, 1997, 334, 1-23). These examples do not preclude the use of other possible cell expression systems, including cell lines obtained from nematodes (PCT application WO 98/37177).
  • An expressed phosphatase can be used for HTS binding assays in conjunction with its defined ligand, in this case the corresponding peptide that activates it. The identified peptide is labeled with a suitable radioisotope, including, but not limited to, [0100] 125I, 3H, 35S or 32P, by methods that are well known to those skilled in the art. Alternatively, the peptides may be labeled by well-known methods with a suitable fluorescent derivative (Baindur, et al., Drug Dev. Res., 1994, 33, 373-398; Rogers, Drug Discovery Today, 1997, 2, 156-160). Radioactive ligand specifically bound to the receptor in membrane preparations made from the cell line expressing the recombinant protein can be detected in HTS assays in one of several standard ways, including filtration of the receptor-ligand complex to separate bound ligand from unbound ligand (Williams, Med. Res. Rev., 1991, 11, 147-184.; Sweetnam, et al., J. Natural Products, 1993, 56, 441-455). Alternative methods include a scintillation proximity assay (SPA) or a FlashPlate format in which such separation is unnecessary (Nakayama, Cur. Opinion Drug Disc. Dev., 1998, 1, 85-91 Bossé, et al., J. Biomolecular Screening, 1998, 3, 285-292.). Binding of fluorescent ligands can be detected in various ways, including fluorescence energy transfer (FRET), direct spectrophotofluorometric analysis of bound ligand, or fluorescence polarization (Rogers, Drug Discovery Today, 1997, 2, 156-160; Hill, Cur. Opinion Drug Disc. Dev., 1998, 1, 92-97).
  • The phosphatases and natural binding partners required for functional expression of heterologous phosphatase polypeptides can be native constituents of the host cell or can be introduced through well-known recombinant technology. The phosphatase polypeptides can be intact or chimeric. The phosphatase activation results in the stimulation or inhibition of other native proteins, events that can be linked to a measurable response. [0101]
  • Examples of such biological responses include, but are not limited to, the following: the ability to survive in the absence of a limiting nutrient in specifically engineered yeast cells (Pausch, [0102] Trends in Biotechnology, 1997, 15, 487-494); changes in intracellular Ca2+ concentration as measured by fluorescent dyes (Murphy, et al., Cur. Opinion Drug Disc. Dev., 1998, 1, 192-199). Fluorescence changes can also be used to monitor ligand-induced changes in membrane potential or intracellular pH; an automated system suitable for HTS has been described for these purposes (Schroeder, et al., J. Biomolecular Screening, 1996, 1, 75-80). Assays are also available for the measurement of common second but these are not generally preferred for HTS.
  • The invention contemplates a multitude of assays to screen and identify inhibitors of ligand binding to phosphatase polypeptides. In one example, the phosphatase polypeptide is immobilized and interaction with a binding partner is assessed in the presence and absence of a candidate modulator such as an inhibitor compound. In another example, interaction between the phosphatase polypeptide and its binding partner is assessed in a solution assay, both in the presence and absence of a candidate inhibitor compound. In either assay, an inhibitor is identified as a compound that decreases binding between the phosphatase polypeptide and its natural binding partner. Another contemplated assay involves a variation of the di-hybrid assay wherein an inhibitor of protein/protein interactions is identified by detection of a positive signal in a transformed or transfected host cell, as described in PCT publication number WO 95/20652, published Aug. 3, 1995 and is included by reference herein including any figures, tables, or drawings. [0103]
  • Candidate modulators contemplated by the invention include compounds selected from libraries of either potential activators or potential inhibitors. There are a number of different libraries used for the identification of small molecule modulators, including: (1) chemical libraries, (2) natural product libraries, and (3) combinatorial libraries comprised of random peptides, oligonucleotides or organic molecules. Chemical libraries consist of random chemical structures, some of which are analogs of known compounds or analogs of compounds that have been identified as “hits” or “leads” in other drug discovery screens, while others are derived from natural products, and still others arise from non-directed synthetic organic chemistry. Natural product libraries are collections of microorganisms, animals, plants, or marine organisms which are used to create mixtures for screening by: (1) fermentation and extraction of broths from soil, plant or marine microorganisms or (2) extraction of plants or marine organisms. Natural product libraries include polyketides, non-ribosomal peptides, and variants (non-naturally occurring) thereof. For a review, see [0104] Science 282:63-68 (1998). Combinatorial libraries are composed of large numbers of peptides, oligonucleotides, or organic compounds as a mixture. These libraries are relatively easy to prepare by traditional automated synthesis methods, PCR, cloning, or proprietary synthetic methods. Of particular interest are non-peptide combinatorial libraries. Still other libraries of interest include peptide, protein, peptidomimetic, multiparallel synthetic collection, recombinatorial, and polypeptide libraries. For a review of combinatorial chemistry and libraries created therefrom, see Myers, Curr. Opin. Biotechnol. 8:701-707 (1997). Identification of modulators through use of the various libraries described herein permits modification of the candidate “hit” (or “lead”) to optimize the capacity of the “hit” to modulate activity.
  • Still other candidate inhibitors contemplated by the invention can be designed and include soluble forms of binding partners, as well as such binding partners as chimeric, or fusion, proteins. A “binding partner” as used herein broadly encompasses both natural binding partners as described above as well as chimeric polypeptides, peptide modulators other than natural ligands, antibodies, antibody fragments, and modified compounds comprising antibody domains that are immunospecific for the expression product of the identified phosphatase gene. [0105]
  • Other assays may be used to identify specific peptide ligands of a phosphatase polypeptide, including assays that identify ligands of the target protein through measuring direct binding of test ligands to the target protein, as well as assays that identify ligands of target proteins through affinity ultrafiltration with ion spray mass spectroscopy/HPLC methods or other physical and analytical methods. Alternatively, such binding interactions are evaluated indirectly using the yeast two-hybrid system described in Fields et al., [0106] Nature, 340:245-246 (1989), and Fields et al., Trends in Genetics, 10:286-292 (1994), both of which are incorporated herein by reference. The two-hybrid system is a genetic assay for detecting interactions between two proteins or polypeptides. It can be used to identify proteins that bind to a known protein of interest, or to delineate domains or residues critical for an interaction. Variations on this methodology have been developed to clone genes that encode DNA binding proteins, to identify peptides that bind to a protein, and to screen for drugs. The two-hybrid system exploits the ability of a pair of interacting proteins to bring a transcription activation domain into close proximity with a DNA binding domain that binds to an upstream activation sequence (UAS) of a reporter gene, and is generally performed in yeast. The assay requires the construction of two hybrid genes encoding (1) a DNA-binding domain that is fused to a first protein and (2) an activation domain fused to a second protein. The DNA-binding domain targets the first hybrid protein to the UAS of the reporter gene; however, because most proteins lack an activation domain, this DNA-binding hybrid protein does not activate transcription of the reporter gene. The second hybrid protein, which contains the activation domain, cannot by itself activate expression of the reporter gene because it does not bind the UAS. However, when both hybrid proteins are present, the noncovalent interaction of the first and second proteins tethers the activation domain to the UAS, activating transcription of the reporter gene. For example, when the first protein is a phosphatase gene product, or fragment thereof, that is known to interact with another protein or nucleic acid, this assay can be used to detect agents that interfere with the binding interaction. Expression of the reporter gene is monitored as different test agents are added to the system. The presence of an inhibitory agent results in lack of a reporter signal.
  • When the function of the phosphatase polypeptide gene product is unknown and no ligands are known to bind the gene product, the yeast two-hybrid assay can also be used to identify proteins that bind to the gene product. In an assay to identify proteins that bind to a phosphatase polypeptide, or fragment thereof, a fusion polynucleotide encoding both a phosphatase polypeptide (or fragment) and a UAS binding domain (i.e., a first protein) may be used. In addition, a large number of hybrid genes each encoding a different second protein fused to an activation domain are produced and screened in the assay. Typically, the second protein is encoded by one or more members of a total-cDNA or genomic DNA fusion library, with each second protein coding region being fused to the activation domain. This system is applicable to a wide variety of proteins, and it is not even necessary to know the identity or function of the second binding protein. The system is highly sensitive and can detect interactions not revealed by other methods; even transient interactions may trigger transcription to produce a stable mRNA that can be repeatedly translated to yield the reporter protein. [0107]
  • Other assays may be used to search for agents that bind to the target protein. One such screening method to identify direct binding of test ligands to a target protein is described in U.S. Pat. No. 5,585,277, incorporated herein by reference. This method relies on the principle that proteins generally exist as a mixture of folded and unfolded states, and continually alternate between the two states. When a test ligand binds to the folded form of a target protein (i.e., when the test ligand is a ligand of the target protein), the target protein molecule bound by the ligand remains in its folded state. Thus, the folded target protein is present to a greater extent in the presence of a test ligand which binds the target protein, than in the absence of a ligand. Binding of the ligand to the target protein can be determined by any method which distinguishes between the folded and unfolded states of the target protein. The function of the target protein need not be known in order for this assay to be performed. Virtually any agent can be assessed by this method as a test ligand, including, but not limited to, metals, polypeptides, proteins, lipids, polysaccharides, polynucleotides and small organic molecules. [0108]
  • Another method for identifying ligands of a target protein is described in Wieboldt et al., [0109] Anal. Chem., 69:1683-1691 (1997), incorporated herein by reference. This technique screens combinatorial libraries of 20-30 agents at a time in solution phase for binding to the target protein. Agents that bind to the target protein are separated from other library components by simple membrane washing. The specifically selected molecules that are retained on the filter are subsequently liberated from the target protein and analyzed by HPLC and pneumatically assisted electrospray (ion spray) ionization mass spectroscopy. This procedure selects library components with the greatest affinity for the target protein, and is particularly useful for small molecule libraries.
  • In preferred embodiments of the invention, methods of screening for compounds which modulate phosphatase activity comprise contacting test compounds with phosphatase polypeptides and assaying for the presence of a complex between the compound and the phosphatase polypeptide. In such assays, the ligand is typically labelled. After suitable incubation, free ligand is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of the particular compound to bind to the phosphatase polypeptide. [0110]
  • In another embodiment of the invention, high throughput screening for compounds having suitable binding affinity to phosphatase polypeptides is employed. Briefly, large numbers of different small peptide test compounds are synthesised on a solid substrate. The peptide test compounds are contacted with the phosphatase polypeptide and washed. Bound phosphatase polypeptide is then detected by methods well known in the art. Purified polypeptides of the invention can also be coated directly onto plates for use in the aforementioned drug screening techniques. In addition, non-neutralizing antibodies can be used to capture the protein and immobilize it on the solid support. [0111]
  • Other embodiments of the invention comprise using competitive screening assays in which neutralizing antibodies capable of binding a polypeptide of the invention specifically compete with a test compound for binding to the polypeptide. In this manner, the antibodies can be used to detect the presence of any peptide that shares one or more antigenic determinants with a phosphatase polypeptide. Radiolabeled competitive binding studies are described in A. H. Lin et al. [0112] Antimicrobial Agents and Chemotherapy, 1997, vol. 41, no. 10. pp. 2127-2131, the disclosure of which is incorporated herein by reference in its entirety.
  • In another aspect, the invention provides methods for treating a disease by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. Preferably the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders. More specifically these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplant rejection. [0113]
  • In preferred embodiments, the invention provides methods for treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. Preferably the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders. More specifically these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplant rejection. [0114]
  • The invention also features methods of treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. Preferably the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders. More specifically these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial- organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplant rejection. [0115]
  • The invention also features methods of treating or preventing a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase polypeptide having an amino acid sequence selected from the group consisting those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24. Preferably the disease is selected from the group consisting of immune-related diseases and disorders, cardiovascular disease, and cancer. Most preferably, the immune-related diseases and disorders are selected from the group consisting of rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplantation. [0116]
  • Substances useful for treatment of phosphatase-related disorders or diseases preferably show positive results in one or more in vitro assays for an activity corresponding to treatment of the disease or disorder in question (Examples of such assays are provided and referenced herein). Examples of substances that can be screened for favorable activity are provided and referenced below. The substances that modulate the activity of the phosphatases preferably include, but are not limited to, antisense oligonucleotides and inhibitors of protein phosphatases, as determined by methods and screens referenced below. [0117]
  • The term “preventing” refers to decreasing the probability that an organism contracts or develops an abnormal condition. [0118]
  • The term “treating” refers to having a therapeutic effect and at least partially alleviating or abrogating an abnormal condition in the organism. [0119]
  • The term “therapeutic effect” refers to the inhibition or activation factors causing or contributing to the abnormal condition. A therapeutic effect relieves to some extent one or more of the symptoms of the abnormal condition. In reference to the treatment of abnormal conditions, a therapeutic effect can refer to one or more of the following: (a) an increase in the proliferation, growth, and/or differentiation of cells; (b) inhibition (i.e., slowing or stopping) of cell death; (c) inhibition of degeneration; (d) relieving to some extent one or more of the symptoms associated with the abnormal condition; and (e) enhancing the function of the affected population of cells. Compounds demonstrating efficacy against abnormal conditions can be identified as described herein. [0120]
  • The term “abnormal condition” refers to a function in the cells or tissues of an organism that deviates from their normal functions in that organism. An abnormal condition can relate to cell proliferation, cell differentiation, or cell survival. [0121]
  • Abnormal cell proliferative conditions include cancers such as fibrotic and mesangial disorders, abnormal angiogenesis and vasculogenesis, wound healing, psoriasis, diabetes mellitus, and inflammation. [0122]
  • Abnormal differentiation conditions include, but are not limited to neurodegenerative disorders, slow wound healing rates, and slow tissue grafting healing rates. [0123]
  • Abnormal cell survival conditions relate to conditions in which programmed cell death (apoptosis) pathways are activated or abrogated. A number of protein phosphatases are associated with the apoptosis pathways. Aberrations in the function of any one of the protein phosphatases could lead to cell immortality or premature cell death. [0124]
  • The term “aberration”, in conjunction with the function of a phosphatase in a signal transduction process, refers to a phosphatase that is over- or under-expressed in an organism, mutated such that its catalytic activity is lower or higher than wild-type protein phosphatase activity, mutated such that it can no longer interact with a natural binding partner, is no longer modified by another protein phosphatase or protein phosphatase, or no longer interacts with a natural binding partner. [0125]
  • The term “administering” relates to a method of incorporating a compound into cells or tissues of an organism. The abnormal condition can be prevented or treated when the cells or tissues of the organism exist within the organism or outside of the organism. Cells existing outside the organism can be maintained or grown in cell culture dishes. For cells harbored within the organism, many techniques exist in the art to administer compounds, including (but not limited to) oral, parenteral, dermal, injection, and aerosol applications. For cells outside of the organism, multiple techniques exist in the art to administer the compounds, including (but not limited to) cell microinjection techniques, transformation techniques, and carrier techniques. [0126]
  • The abnormal condition can also be prevented or treated by administering a compound to a group of cells having an aberration in a signal transduction pathway to an organism. The effect of administering a compound on organism function can then be monitored. The organism is preferably a mouse, rat, rabbit, guinea pig, or goat, more preferably a monkey or ape, and most preferably a human. [0127]
  • In another aspect, the invention features methods for detection of a phosphatase polypeptide in a sample as a diagnostic tool for diseases or disorders, wherein the method comprises the steps of: (a) contacting the sample with a nucleic acid probe which hybridizes under hybridization assay conditions to a nucleic acid target region of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, said probe comprising the nucleic acid sequence encoding the polypeptide, fragments thereof, and the complements of the sequences and fragments; and (b) detecting the presence or amount of the probe:target region hybrid as an indication of the disease. [0128]
  • In preferred embodiments of the invention, the disease or disorder is selected from the group consisting of rheumatoid arthritis, arteriosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure, oxidative stress-related neurodegenerative disorders, and cancer. [0129]
  • The phosphatase “target region” is the nucleotide base sequence selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or the corresponding full-length sequences, a functional derivative thereof, or a fragment thereof to which the nucleic acid probe will specifically hybridize. Specific hybridization indicates that in the presence of other nucleic acids the probe only hybridizes detectably with the phosphatase of the invention's target region. Putative target regions can be identified by methods well known in the art consisting of alignment and comparison of the most closely related sequences in the database. [0130]
  • In preferred embodiments the nucleic acid probe hybridizes to a phosphatase target region encoding at least 6, 12, 75, 90, 105, 120, 150, 200, 250, 300 or 350 contiguous amino acids of a sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or the corresponding full-length amino acid sequence, or a functional derivative thereof. Hybridization conditions should be such that hybridization occurs only with the phosphatase genes in the presence of other nucleic acid molecules. Under stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having more than 1 or 2 mismatches out of 20 contiguous nucleotides. Such conditions are defined, above. [0131]
  • The diseases for which detection of phosphatase genes in a sample could be diagnostic include diseases in which phosphatase nucleic acid (DNA and/or RNA) is amplified in comparison to normal cells. By “amplification” is meant increased numbers of phosphatase DNA or RNA in a cell compared with normal cells. In normal cells, phosphatases are typically found as single copy genes. In selected diseases, the chromosomal location of the phosphatase genes may be amplified, resulting in multiple copies of the gene, or amplification. Gene amplification can lead to amplification of phosphatase RNA, or phosphatase RNA can be amplified in the absence of phosphatase DNA amplification. [0132]
  • “Amplification” as it refers to RNA can be the detectable presence of phosphatase RNA in cells, since in some normal cells there is no basal expression of phosphatase RNA. In other normal cells, a basal level of expression of phosphatase exists, therefore in these cases amplification is the detection of at least 1-2-fold, and preferably more, phosphatase RNA, compared to the basal level. [0133]
  • The diseases that could be diagnosed by detection of phosphatase nucleic acid in a sample preferably include cancers. The test samples suitable for nucleic acid probing methods of the present invention include, for example, cells or nucleic acid extracts of cells, or biological fluids. The samples used in the above-described methods will vary based on the assay format, the detection method and the nature of the tissues, cells or extracts to be assayed. Methods for preparing nucleic acid extracts of cells are well known in the art and can be readily adapted in order to obtain a sample that is compatible with the method utilized. [0134]
  • In another aspect, the invention features a method for detection of a phosphatase polypeptide in a sample as a diagnostic tool for a disease or disorder, wherein the method comprises: (a) comparing a nucleic acid target region encoding the phosphatase polypeptide in a sample, where the phosphatase polypeptide has an amino acid sequence selected from the group consisting those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or one or more fragments thereof, with a control nucleic acid target region encoding the phosphatase polypeptide, or one or more fragments thereof; and (b) detecting differences in sequence or amount between the target region and the control target region, as an indication of the disease or disorder. Preferably the disease is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders. More specifically these diseases include cancer of tissues or hematopoietic origin; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, Multiple sclerosis, and Amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplant rejection. [0135]
  • The term “comparing” as used herein refers to identifying discrepancies between the nucleic acid target region isolated from a sample, and the control nucleic acid target region. The discrepancies can be in the nucleotide sequences, e.g. insertions, deletions, or point mutations, or in the amount of a given nucleotide sequence. Methods to determine these discrepancies in sequences are well-known to one of ordinary skill in the art. The “control” nucleic acid target region refers to the sequence or amount of the sequence found in normal cells, e.g. cells that are not diseased as discussed previously. [0136]
  • Method of Use [0137]
  • Partial amino sequences for human protein phosphatases are encoded by nucleic acid sequences set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12. [0138]
  • These sequences will be used to find the full-length clone of each of the predicted protein phosphatases. These clones will be useful for screening for small molecule compounds that inhibit the catalytic activity of the encoded protein phosphatase with potential utility in treating disorders including cancers of tissues or blood particular those involving breast, colon, lung, prostate, cervical, brain, ovarian, bladder, or kidney; central or peripheral nervous system diseases and conditions including migraine, pain, sexual dysfunction, mood disorders, attention disorders, cognition disorders, hypotension, and hypertension; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, delirium, dementia, severe mental retardation and dyskinesias, such as Huntington's disease or Tourette's Syndrome; neurodegenerative diseases including Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis; viral infections caused by HIV-1, HIV-2 or other viral- or prion-agents or fungal- or bacterial-organisms; metabolic disorders including Diabetes and obesity and their related syndromes, among others; cardiovascular disorders including reperfusion restenosis, coronary thrombosis, clotting disorders, unregulated cell growth disorders, atherosclerosis; ocular disease including glaucoma, retinopathy, and macular degeneration; inflammatory disorders including rheumatoid arthritis, chronic inflammatory bowel disease, chronic inflammatory pelvic disease, multiple sclerosis, asthma, osteoarthritis, psoriasis, atherosclerosis, rhinitis, autoimmunity, and organ transplant rejection. [0139]
  • The summary of the invention described above is not limiting and other features and advantages of the invention will be apparent from the following detailed description of the invention, and from the claims.[0140]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. [0141] 1A-H show the nucleotide sequences for human protein phosphatases (SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12).
  • FIGS. [0142] 2A-2C provide amino acid sequences for the human protein phosphatases encoded by SEQ ID NO: 1-NO: 12 (SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, respectively). Some of the sequences encode predicted stop codons within the coding region, indicated by an ‘x.’
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the isolation and characterization of new polypeptides, nucleotide sequences encoding these polypeptides, various products and assay methods that can be used to identify compounds useful for the diagnosis and treatment of various polypeptide-related diseases and conditions, for example cancer. Polypeptides, preferably phosphatases, and nucleic acids encoding such polypeptides may be produced, using well-known and standard synthesis techniques when given the sequences presented herein. By reference, e.g., to Tables 1 though 8, below, genes according to the invention can be better understood. The invention additionally provides a number of different embodiments, such as those described below. [0143]
  • Nucleic Acids [0144]
  • Associations of chromosomal localizations for mapped genes with amplicons implicated in cancer are based on literature searches (PubMed http://www.ncbi.nlm.nih.gov/entrez/query.fcgi), OMIM searches (Online Mendelian Inheritance in Man, http://www.ncbi.nlm.nih.gov/Omim/searchomim.html) and the comprehensive database of cancer amplicons maintained by Knuutila, et al. (Knuutila, et al., [0145] DNA copy number amplifications in human neoplasms. Review of comparative genomic hybridization studies. Am J Pathol 152:1107-1123, 1998. http://www.helsinki.fi/˜lgl_www/CMG.html). For many of the mapped genes, the cytogenetic region from Knuutila is listed followed by the number of cases with documented amplification and the total number of cases studied. Thus for SGP006 below, the entry “Bladder carcinoma (12q21-q24, 1/16)” means that the chromosomal position has been associated with non-small cell lung cancer, at position 12q21-q24, which encompasses the SGP006's position, and the amplification has been noted in 1 of the 16 samples studied.
  • For single nucleotide polymorphisms, an accession number (for example, ss1581624 for SGP187) is given if the SNP is documented in dbSNP (the database of single nucleotide polymorphisms) maintained at NCBI (http://www.ncbi.nh.nih.gov/SNP/index.html). The accession number for SNP can be used to retrieve the full SNP-containing sequence from this site. Candidate SNPs without a dbSNP accession number were identified by inspection of Blastn outputs of the patent sequences vs cDNA and genomic databases, as shown in Table 7 and Table 8, respectively, in Example 1. [0146]
  • Nucleic Acid Probes, Methods, and Kits for Detection of Phosphatases [0147]
  • The present invention additionally provides nucleic acid probes an uses therefor. A nucleic acid probe of the present invention may be used to probe an appropriate chromosomal or cDNA library by usual hybridization methods to obtain other nucleic acid molecules of the present invention. A chromosomal DNA or cDNA library may be prepared from appropriate cells according to recognized methods in the art (cf. “Molecular Cloning: A Laboratory Manual”, second edition, Cold Spring Harbor Laboratory, Sambrool, Fritsch, & Maniatis, eds., 1989). [0148]
  • In the alternative, chemical synthesis can be carried out in order to obtain nucleic acid probes having nucleotide sequences which correspond to N-terminal and C-terminal portions of the amino acid sequence of the polypeptide of interest. The synthesized nucleic acid probes may be used as primers in a polymerase chain reaction (PCR) carried out in accordance with recognized PCR techniques, essentially according to PCR Protocols, “A Guide to Methods and Applications”, Academic Press, Michael, et al., eds., 1990, utilizing the appropriate chromosomal or cDNA library to obtain the fragment of the present invention. [0149]
  • One skilled in the art can readily design such probes, based on the nucleic acid and amino acid sequences disclosed herein, using methods of computer alignment and sequence analysis known in the art (“Molecular Cloning: A Laboratory Manual”, 1989, supra). The hybridization probes of the present invention can be labeled by standard labeling techniques such as with a radiolabel, enzyme label, fluorescent label, biotin-avidin label, chemiluminescence, and the like. After hybridization, the probes may be visualized using known methods. [0150]
  • The nucleic acid probes of the present invention include RNA, as well as DNA probes, such probes being generated using techniques known in the art. The nucleic acid probe may be immobilized on a solid support. Examples of such solid supports include, but are not limited to, plastics such as polycarbonate, complex carbohydrates such as agarose and sepharose, and acrylic resins, such as polyacrylamide and latex beads. Techniques for coupling nucleic acid probes to such solid supports are well known in the art. [0151]
  • The test samples suitable for nucleic acid probing methods of the present invention include, for example, cells or nucleic acid extracts of cells, or biological fluids. The samples used in the above-described methods will vary based on the assay format, the detection method and the nature of the tissues, cells or extracts to be assayed. Methods for preparing nucleic acid extracts of cells are well known in the art and can be readily adapted in order to obtain a sample which is compatible with the method utilized. [0152]
  • One method of detecting the presence of nucleic acids of the invention in a sample comprises (a) contacting said sample with the above-described nucleic acid probe under conditions such that hybridization occurs, and (b) detecting the presence of said probe bound to said nucleic acid molecule. One skilled in the art would select the nucleic acid probe according to techniques known in the art as described above. Samples to be tested include but should not be limited to RNA samples of human tissue. [0153]
  • A kit for detecting the presence of nucleic acids of the invention in a sample comprises at least one container means having disposed therein the above-described nucleic acid probe. The kit may further comprise other containers comprising one or more of the following: wash reagents and reagents capable of detecting the presence of bound nucleic acid probe. Examples of detection reagents include, but are not limited to radiolabelled probes, enzymatic labeled probes (horseradish peroxidase, alkaline phosphatase), and affinity labeled probes (biotin, avidin, or steptavidin). Preferably, the kit further comprises instructions for use. [0154]
  • In detail, a compartmentalized kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers or strips of plastic or paper. Such containers allow the efficient transfer of reagents from one compartment to another compartment such that the samples and reagents are not cross-contaminated and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another. Such containers will include a container which will accept the test sample, a container which contains the probe or primers used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, and the like), and containers which contain the reagents used to detect the hybridized probe, bound antibody, amplified product, or the like. One skilled in the art will readily recognize that the nucleic acid probes described in the present invention can readily be incorporated into one of the established kit formats which are well known in the art. [0155]
  • Categorization of the Polypeptides According to the Invention [0156]
  • For a number of protein phosphatases of the invention, there is provided a classification of the protein class and family to which it belongs, a summary of non-catalytic protein motifs, as well as a chromosomal location. This information is useful in determing function, regulation and/or therapeutic utility for each of the proteins. Amplification of chromosomal region can be associated with various cancers. For amplicons discussed in this application, the source of information was Knuutila, et al (Knuutila S, Björkqvist A-M, Autio K, Tarkkanen M, Wolf M, Monni O, Szymanska J, Larramendy M L, Tapper J, Pere H, El-Rifai W, Hemmer S, Wasenius V-M, Vidgren V & Zhu Y: DNA copy number amplifications in human neoplasms. Review of comparative genomic hybridization studies. Am J Pathol 152:1107-1123, 1998. http://www.helsinld.fi/˜lgl_www/CMG.html). [0157]
  • The phosphatase classification and protein domains often reflect pathways, cellular roles, or mechanisms of up- or down-stream regulation. Also disease-relevant genes often occur in families of related genes. For example, if one member of a phosphatase family functions as an oncogene, a tumor suppressor, or has been found to be disrupted in an immune, neurologic, cardiovascular, or metabolic disorder, frequently other family members may play a related role. [0158]
  • Chromosomal location can identify candidate targets for a tumor amplicon or a tumor-suppressor locus. Summaries of prevalent tumor amplicons are available in the literature, and can identify tumor types to experimentally be confirmed to contain amplified copies of a phosphatase gene which localizes to an adjacent region. [0159]
  • A more specific characterization of the polypeptides of the invention, including potential biological and clinical implications, is provided, e.g., in EXAMPLES 2 and 3. [0160]
  • Classification of Polypeptides Exhibiting Phosphatase Activity [0161]
  • The polypeptides described in the present invention may belong to one of the following groups: (1) dual-specificity group of protein phosphatases (DSP); (2) serine-threonine phosphatases (STP); or (3) protein tyrosine phosphatases (PTP). This classification relies, at least in part, on the conserved core amino acid sequence motifs that make up the catalytic domain of this class of phosphatases. [0162]
  • DSP Group [0163]
  • The unique signature motifs of the catalytic domain of the dual-specificity class of phosphatases is responsible for the ability of these enzymes to dephosphorylate phosphoserine/phosphothreonine as well phosphotyrosine residues. The dual-specificity group of protein phosphatases include the family member MAP kinase phosphatases (MKP). A description of the structural and functional characteristics for the MKP family now follows. [0164]
  • MKP Family [0165]
  • Novel MKP-like phosphatases in this application include SGP006 (SEQ ID NO: 1), SGP002 (SEQ ID NO: 2), SGP001 (SEQ ID NO: 3), SGP018 (SEQ ID NO: 4), SGP003 (SEQ ID NO: 5), SGP014 (SEQ ID NO: 6), SGP060 (SEQ ID NO: 7), and SGP008 (SEQ ID NO: 8), which are disclosed in greater detail in the Tables 1-6 and Example 2, for example. [0166]
  • The dual specificity phosphatase family includes around 20 known human members (for a list, see http://smart.embl-heidelberg.de/smart/get_members.pl?WHAT=species&NAME=DSPc&WHICH=Ho mo_sapiens). Well-known members of the MPK family of dual-specificity phosphatases include: DUS1 (also known as MPK-1, CL100, PTPN-10, erp, VH1 or 3CH134), DUS3 (also known as VHR), DUS4 (also known as HVH2, TYP1, MKP2 or VH2), DUS5 (also known as HVH3, B23, VH3), DUS6 (also known as PYST1, MKP3, rVH6), DUS7 (also known as PYST2), CDKN3 (also known as CDKN3, KAP, CIP2 or CDI1), VH5 and STYX. [0167]
  • Most MKP phosphatases are capable of inactivating, through a dephosphorylation reaction, kinases that participate in the MAPK pathways. The ERK (extracellular signal-regulated kinase), JNK/SAPK (c-Jun N-terminal kinase/stress-activated protein kinase) and p38 MAP kinase pathways mediate the signal transduction events that are responsible for cell division, differentiation or apoptosis in response to extracellular ligands (Cobb M H, Prog Biophys Mol Biol. 1999;71(3-4):479-500). Full MAP kinase enzymatic activation requires the concomitant phosphorylation by selective upstream dual-specificity kinases of threonine and tyrosine residues residing in the activation loop of the MAP kinases. MKP family dual-specificity phosphatases mediate MAP kinase inactivation by dephosphorylating these threonine and tyrosine residues. This mechanism provides negative feedback regulation of the MAP kinase pathways. MKPs may play a significant role in human cancer by attenuating MAP kinase cascades involved in cellular transformation. [0168]
  • Given the large number of MAP kinases, as well as MKP's, a central question is whether there is selectivity in kinase substrate recognition by MKP's. Evidence that such specificity exists is provided by DUS-6 (MKP3) and VH5 which have been shown to be highly selective phosphatases towards the ERK or JNK/SAPK and p38 MAP kinases, respectively (Muda M, et al., J Biol Chem. Nov. 1, 1996;271(44):27205-8.). Another level of substrate specificity comes from subcellular compartmentalization as shown by DUS-6 (MKP3) which is found exclusively in the cytosol rather than in the nucleus (Groom, L. A. et al (1996) EMBO J. 15: 3621-3632). Further specificity can arise at the level of the tissue specificity of expression (i.e. Muda, M. et al (1997) J. Biol. Chem. 272:5141-5151). [0169]
  • MKP's appear to be as ubiquitous in their phylogenetic distribution as their MAP kinase counterparts with multiple members present in yeast (i.e. YVH1), [0170] C. elegans (i.e. Y042), Drosophila, (i.e. puckered), plants (i.e. DsPTP1) and mammals. The primary mode of action of MKP's isolated from different species appears to be MAPK dephosphorylation thereby providing negative feedback to the MAPK signal transduction pathways.
  • MKP's may play an important role during pathophysiological hypoxia as suggested by the induction of MKP-1 gene expression under low oxygen conditions (Laderroute, K. R. (1999) J. Biol. Chem. 274:12890-12897). Tumor hypoxia is directly linked to the onset of angiogenesis during malignant progression (Hanahan, D. et al (1996) Cell 86:353-364 and Mazure, N. M. et al (1996) Cancer Res. 56:3436-3440). A number of genes have been found to be induced during hypoxic conditions such as the heat shock transcription factor-1 (HSF-1) (Benjamin, I. J. et al. (1990) Proc. Natl. Acad. Sci. 87:6263-6267), c-fos and c-jun (Ausserer, W. A. et al (1994) Mol. Cell. Biol. 14:5032-5042, and Muller, J. M. (1997) J. Biol. Chem 272:23435-23439) and the hypoxia-inducible factor-1 (HIF-1) (Wenger, R. H. et al (1997) J. Biol. Chem. 378:609-616). MKP-1 transcripts and protein have been shown to be upregulated in early-stage carcinomas well as in multiple stages of breast and prostate carcinomas (i.e. Leav, I. Et al (1996) Lab. Invest. 75: 361-370). Over-expression of MKP-1 in prostate tumor cell lines confers resistance to Fas ligand-induced apoptosis (Srikanth, S. et al. (1999) Mol. Cell. Biochem. 199: 169-178) and it has also been suggested that MKP-1 may contribute to the inhibition of apoptosis resulting in androgen-independent growth. MKP-1 may also inhibit the induction of apoptosis that is produced by anti-neoplastic agents such as cisplatin and camptothecin (Sanchez-Perez, I et al. (2000) Oncogene 19: 5142-5152; Costa-Pereira, A. P. et al. (2000) Br. J. Cancer 82: 1827-1834). Since hypoxic conditions are known to trigger apoptosis via the activation of the JNK pathway (reviewed in Ip, Y. T. et al (1998) Curr. Opin. Cell Biol. 10:205-219) and MAPK phosphatases provide negative feedback to this pathway, it is conceivable that MKP-1 supports tumor growth by blocking apoptosis. Over-expression of MKP-1 can block the hypoxia-induced activation of SAPK/JNK in co-transfected tumor cells (Laderroute, K. R. (1999) J. Biol. Chem. 274:12890-12897). [0171]
  • The dephosphorylation and subsequent inactivation of ERK-1 and ERK-2 by MAPK phosphatases may also be responsible for suppressing angiogenic vascular endothelial cell proliferation by angiostatin Redlitz, A. et al. (1999 J. Vasc. Res 36:28-34). [0172]
  • The novel MPK family phosphatases presented in this filing contribute to a growing list of phosphatases that appear to have as their primary function negative feedback regulation of MAPK signal transduction. Since there is precedence for selectivity in the mechanism of action at the level of substrate recognition, subcellular localization and tissue distribution among the known MPK's, the novel MPK's described may display similar selectivity. The novel MPK's may also play a role in suppressing apoptosis by blocking the JNK/SAPK pathway during pathological hypoxia such as that occurring in angiogenic tumors. The development of specific phosphatase inhibitors that target the anti-apoptotic MKP's may prove valuable as an approach to cancer therapy. [0173]
  • PTP Group [0174]
  • There are 2 PTP-like sequences in this application: SGP012 (SEQ ID NO: 11) and SGP024 (SEQ ID NO: 12), which are disclosed in greater detail in the Tables 1-6 and Example 2, for example. [0175]
  • SGP012 is closely related to murine OST-PTP, also called PTP-ESP. Osteotesticular PTP (OST-PTP) is a putative receptor protein tyrosine phosphatase that possesses 10 fibronectin type m repeats, a potential membrane-spanning region and an intracellular domain consisting of two tandem catalytic domains. The expression pattern is highly restricted and is detectable primarily in bone and testis (Mauro et al. J. Biol Chem 1996 269:30659-67). The ligand for OST-PTP is not known but the structure of the extracellular domain suggests that cell-cell interactions may be involved. Importantly, the human ortholog has not yet been cloned. [0176]
  • The balance between bone deposition and resorption is controlled by the relative activities of two cell types, osteoblasts and osteoclasts. The potential role of phosphatases in bone metabolism is only incompletely understood. However, in osteoblast cultures, inhibition of PTP activity with orthovanadate enhances matrix formation (Lau et al. Endocrinology 188 123:2858-67). In addition, bisphophonates, which are used clinically to treat bone diseases with excess resorption, cause a range of changes in osteoblast cultures that are consistent with increased bone deposition including osteoblast differentiation, alkaline phosphatase activity, type I collagen secretion, and mineralization (Reinholz et al. Cancer Research 2000 60:6001-007). The molecular target of these compounds is still unknown, but it is plausible that inhibition of OST-PTP activity is responsible for the observed increases in bone-forming activities in osteoblast cultures. Therefore targeting of OST-PTP activity could provide treatments for osteoporosis, non-healing fractures, and other disorders of bone metabolism. [0177]
  • SGP024 represents a partial PTPT catalytic domain related to PTP-delta. [0178]
  • STP Group [0179]
  • There are 2 STP proteins in this application: SGP039 (SEQ ID NO: 9) and SGP040 (SEQ ID NO: 10), which are disclosed in greater detail in the Tables 1-6 and Example 2, for example. [0180]
  • The Serine-threonine phosphatases can be divided into four major classes represented by PP1, PP2A, PP2B, and PP2C. PP2a is found associated with multiple regulatory subunits and its inactivation leads to transformation by viral components such as small T antigen. Mutations in one of the regulatory subunits have been associated with colorectal cancers consistent with a role as a tumor suppressor (Talcagi et al. Gut 2000 47:268-71. Recently, PP2a has also been implicated in activation of T lymphocytes (Chuang et al. Immunity 2000 13:313-22). PP1 has been implicated in a variety of cellular functions including response to hypoxia, apoptosis and cytokinesis (Taylor et al., PNAS 2000 97:12091-96, Aylion et al. EMBO J 2000 19 2237-46, Orr et al., [0181] Infect. Immun. 2000 68:1350-58). Finally, studies in diabetic rats showed decreased PP1 activity and elevated PP2A activity compared to controls (Begum and Ragolia Metabolism 1998 47:54-62). Because of the diversity of regulatory subunits that affect the activity of serine-threonine phosphatases, biological function of novel members are difficult to predict. However, the studies suggest potential involvement in a variety of diseases including tumorigenesis, inflammatory diseases, and metabolic diseases.
  • Therapeutic Methods According to the Invention: [0182]
  • Diagnostics: [0183]
  • The invention provides methods for detecting a polypeptide in a sample as a diagnostic tool for diseases or disorders, wherein the method comprises the steps of: (a) contacting the sample with a nucleic acid probe which hybridizes under hybridization assay conditions to a nucleic acid target region of a polypeptide selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17; SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, said probe comprising the nucleic acid sequence encoding the polypeptide, fragments thereof, and the complements of the sequences and fragments; and (b) detecting the presence or amount of the probe:target region hybrid as an indication of the disease. [0184]
  • In preferred embodiments of the invention, the disease or disorder is selected from the group consisting of rheumatoid arthritis, atherosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure, oxidative stress-related neurodegenerative disorders, metabolic disorder including diabetes, reproductive disorders including infertility, and cancer. [0185]
  • Hybridization conditions should be such that hybridization occurs only with the genes in the presence of other nucleic acid molecules. Under stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having 1 or 2 mismatches out of 20 contiguous nucleotides. Such conditions are defined herein. [0186]
  • The diseases for which detection of genes in a sample could be diagnostic include diseases in which nucleic acid (DNA and/or RNA) is amplified in comparison to normal cells. By “amplification” is meant increased numbers of DNA or RNA in a cell compared with normal cells. [0187]
  • “Amplification” as it refers to RNA can be the detectable presence of RNA in cells, since in some normal cells there is no basal expression of RNA. In other normal cells, a basal level of expression exists, therefore in these cases amplification is the detection of at least 1-2-fold, and preferably more, compared to the basal level. [0188]
  • The diseases that could be diagnosed by detection of nucleic acid in a sample preferably include cancers. The test samples suitable for nucleic acid probing methods of the present invention include, for example, cells or nucleic acid extracts of cells, or biological fluids. The samples used in the above-described methods will vary based on the assay format, the detection method and the nature of the tissues, cells or extracts to be assayed. Methods for preparing nucleic acid extracts of cells are well known in the art and can be readily adapted in order to obtain a sample that is compatible with the method utilized. [0189]
  • Antibodies, Hybridomas, Methods of Use and Kits for Detection Phosphatases: [0190]
  • The present invention relates to an antibody having binding affinity to a phosphatase of the invention. The polypeptide may have the amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or a functional derivative thereof, or at least 9 contiguous amino acids thereof (preferably, at least 20, 30, 35, or 40 contiguous amino acids thereof). [0191]
  • The present invention also relates to an antibody having specific binding affinity to a phosphatase of the invention. Such an antibody may be isolated by comparing its binding affinity to a phosphatase of the invention with its binding affinity to other polypeptides. Those which bind selectively to a phosphatase of the invention would be chosen for use in methods requiring a distinction between a phosphatase of the invention and other polypeptides. Such methods could include, but should not be limited to, the analysis of altered phosphatase expression in tissue containing other polypeptides. [0192]
  • The phosphatases of the present invention can be used in a variety of procedures and methods, such as for the generation of antibodies, for use in identifying pharmaceutical compositions, and for studying DNA/protein interaction. [0193]
  • The phosphatases of the present invention can be used to produce antibodies or hybridomas. One skilled in the art will recognize that if an antibody is desired, such a peptide could be generated as described herein and used as an immunogen. The antibodies of the present invention include monoclonal and polyclonal antibodies, as well fragments of these antibodies, and humanized forms. Humanized forms of the antibodies of the present invention may be generated using one of the procedures known in the art such as chimerization or CDR grafting. [0194]
  • The present invention also relates to a hybridoma which produces the above-described monoclonal antibody, or binding fragment thereof. A hybridoma is an immortalized cell line which is capable of secreting a specific monoclonal antibody. [0195]
  • In general, techniques for preparing monoclonal antibodies and hybridomas are well known in the art (Campbell, “Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology,” Elsevier Science Publishers, Amsterdam, The Netherlands, 1984; St. Groth et al., J. Immunol. Methods 35:1-21, 1980). Any animal (mouse, rabbit, and the like) which is known to produce antibodies can be immunized with the selected polypeptide. Methods for immunization are well known in the art. Such methods include subcutaneous or intraperitoneal injection of the polypeptide. One skilled in the art will recognize that the amount of polypeptide used for immunization will vary based on the animal which is immunized, the antigenicity of the polypeptide and the site of injection. [0196]
  • The polypeptide may be modified or administered in an adjuvant in order to increase the peptide antigenicity. Methods of increasing the antigenicity of a polypeptide are well known in the art. Such procedures include coupling the antigen with a heterologous protein (such as globulin or β-galactosidase) or through the inclusion of an adjuvant during immunization. [0197]
  • For monoclonal antibodies, spleen cells from the immunized animals are removed, fused with myeloma cells, such as SP2/0-Ag14 myeloma cells, and allowed to become monoclonal antibody producing hybridoma cells. Any one of a number of methods well known in the art can be used to identify the hybridoma cell which produces an antibody with the desired characteristics. These include screening the hybridomas with an ELISA assay, western blot analysis, or radioimmunoassay Lutz et al., Exp. Cell Res. 175:109-124, 1988). Hybridomas secreting the desired antibodies are cloned and the class and subclass are determined using procedures known in the art (Campbell, “Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology”, supra, 1984). [0198]
  • For polyclonal antibodies, antibody-containing antisera is isolated from the immunized animal and is screened for the presence of antibodies with the desired specificity using one of the above-described procedures. The above-described antibodies may be detectably labeled. Antibodies can be detectably labeled through the use of radioisotopes, affinity labels (such as biotin, avidin, and the like), enzymatic labels (such as horseradish peroxidase, alkaline phosphatase, and the like) fluorescent labels (such as FITC or rhodamine, and the like), paramagnetic atoms, and the like. Procedures for accomplishing such labeling are well-known in the art, for example, see Stemberger et al., J. Histochem. Cytochem. 18:315, 1970; Bayer et al., Meth. Enzym. 62:308, 1979; Engval et al., Immunol. 109:129, 1972; Goding, J. Immunol. Meth. 13:215, 1976. The labeled antibodies of the present invention can be used for in vitro, in vivo, and in situ assays to identify cells or tissues which express a specific peptide. [0199]
  • The above-described antibodies may also be immobilized on a solid support. Examples of such solid supports include plastics such as polycarbonate, complex carbohydrates such as agarose and sepharose, acrylic resins such as polyacrylamide and latex beads. Techniques for coupling antibodies to such solid supports are well known in the art (Weir et al., “Handbook of Experimental Immunology” 4th Ed., Blackwell Scientific Publications, Oxford, England, [0200] Chapter 10, 1986; Jacoby et al., Meth. Enzym. 34, Academic Press, N.Y., 1974). The immobilized antibodies of the present invention can be used for in vitro, in vivo, and in situ assays as well as in immunochromotography.
  • Furthermore, one skilled in the art can readily adapt currently available procedures, as well as the techniques, methods and kits disclosed herein with regard to antibodies, to generate peptides capable of binding to a specific peptide sequence in order to generate rationally designed antipeptide peptides (Hurby et al., “Application of Synthetic Peptides: Antisense Peptides”, In Synthetic Peptides, A User's Guide, W. H. Freeman, NY, pp. 289-307, 1992; Kaspczak et al., Biochemistry 28:9230-9238, 1989). [0201]
  • Anti-peptide peptides can be generated by replacing the basic amino acid residues found in the peptide sequences of the phosphatases of the invention with acidic residues, while maintaining hydrophobic and uncharged polar groups. For example, lysine, arginine, and/or histidine residues are replaced with aspartic acid or glutamic acid and glutamic acid residues are replaced by lysine, arginine or histidine. [0202]
  • The present invention also encompasses a method of detecting a phosphatase polypeptide in a sample, comprising: (a) contacting the sample with an above-described antibody, under conditions such that immunocomplexes form, and (b) detecting the presence of said antibody bound to the polypeptide. In detail, the methods comprise incubating a test sample with one or more of the antibodies of the present invention and assaying whether the antibody binds to the test sample. Altered levels of a phosphatase of the invention in a sample as compared to normal levels may indicate disease. [0203]
  • Conditions for incubating an antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the antibody used in the assay. One skilled in the art will recognize that any one of the commonly available immunological assay formats (such as radioimmunoassays, enzyme-linked immunosorbent assays, diffusion-based Ouchterlony, or rocket immunofluorescent assays) can readily be adapted to employ the antibodies of the present invention. Examples of such assays can be found in Chard (“An Introduction to Radioimmunoassay and Related Techniques” Elsevier Science Publishers, Amsterdam, The Netherlands, 1986), Bullock et al. (“Techniques in Immunocytochemistry,” Academic Press, Orlando, Fla. Vol. 1, 1982; Vol. 2, 1983; Vol. 3, 1985), Tijssen (“Practice and Theory of Enzyme Immunoassays: Laboratory Techniques in Biochemistry and Molecular Biology,” Elsevier Science Publishers, Amsterdam, The Netherlands, 1985). [0204]
  • The immunological assay test samples of the present invention include cells, protein or membrane extracts of cells, or biological fluids such as blood, serum, plasma, or urine. The test samples used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed. Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can readily be adapted in order to obtain a sample which is testable with the system utilized. [0205]
  • A kit contains all the necessary reagents to carry out the previously described methods of detection. The kit may comprise: (i) a first container means containing an above-described antibody, and (ii) second container means containing a conjugate comprising a binding partner of the antibody and a label. In another preferred embodiment, the kit further comprises one or more other containers comprising one or more of the following: wash reagents and reagents capable of detecting the presence of bound antibodies. [0206]
  • Examples of detection reagents include, but are not limited to, labeled secondary antibodies, or in the alternative, if the primary antibody is labeled, the chromophoric, enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody. The compartmentalized kit may be as described above for nucleic acid probe kits. One skilled in the art will readily recognize that the antibodies described in the present invention can readily be incorporated into one of the established kit formats which are well known in the art. [0207]
  • Isolation of Compounds Which Interact With Phosphatases [0208]
  • The present invention also relates to a method of detecting a compound capable of binding to a phosphatase of the invention comprising incubating the compound with a phosphatase of the invention and detecting the presence of the compound bound to the phosphatase. The compound may be present within a complex mixture, for example, serum, body fluid, or cell extracts. [0209]
  • The present invention also relates to a method of detecting an agonist or antagonist of phosphatase activity or phosphatase binding partner activity comprising incubating cells that produce a phosphatase of the invention in the presence of a compound and detecting changes in the level of phosphatase activity or phosphatase binding partner activity. The compounds thus identified would produce a change in activity indicative of the presence of the compound. The compound may be present within a complex mixture, for example, serum, body fluid, or cell extracts. Once the compound is identified it can be isolated using techniques well known in the art. [0210]
  • Modulating Polypeptide Activity: [0211]
  • The invention additionally provides methods for treating a disease or abnormal condition by administering to a patient in need of such treatment a substance that modulates the activity of a polypeptide selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, a functional derivative thereof, and a fragment thereof. Preferably, the disease is selected from the group consisting of rheumatoid arthritis, atherosclerosis, autoimmune disorders, organ transplantation, myocardial infarction, cardiomyopathies, stroke, renal failure; oxidative stress-related neurodegenerative disorders, metabolic and reproductive disorders, and cancer. [0212]
  • Substances useful for treatment of disorders or diseases preferably show positive results in one or more assays for an activity corresponding to treatment of the disease or disorder in question Substances that modulate the activity of the polypeptides preferably include, but are not limited to, antisense oligonucleotides and inhibitors of protein phosphatases. [0213]
  • The term “preventing” refers to decreasing the probability that an organism contracts or develops an abnormal condition. [0214]
  • The term “treating” refers to having a therapeutic effect and at least partially alleviating or abrogating an abnormal condition in the organism. [0215]
  • The term “therapeutic effect” refers to the inhibition or activation factors causing or contributing to the abnormal condition. A therapeutic effect relieves to some extent one or more of the symptoms of the abnormal condition. In reference to the treatment of abnormal conditions, a therapeutic effect can refer to one or more of the following: (a) an increase in the proliferation, growth, and/or differentiation of cells; (b) inhibition (, slowing or stopping) of cell death; (c) inhibition of degeneration; (d) relieving to some extent one or more of the symptoms associated with the abnormal condition; and (e) enhancing the function of the affected population of cells. Compounds demonstrating efficacy against abnormal conditions can be identified as described herein. [0216]
  • The term “abnormal condition” refers to a function in the cells or tissues of an organism that deviates from their normal functions in that organism. An abnormal condition can relate to cell proliferation, cell differentiation or cell survival. An abnormal condition may also include irregularities in cell cycle progression, i.e., irregularities in normal cell cycle progression through mitosis and meiosis. [0217]
  • Abnormal cell proliferative conditions include cancers such as fibrotic and mesangial disorders, abnormal angiogenesis and vasculogenesis, wound healing, psoriasis, diabetes mellitus, and inflammation. [0218]
  • Abnormal differentiation conditions include, but are not limited to, neurodegenerative disorders, slow wound healing rates, and slow tissue grafting healing rates. [0219]
  • Abnormal cell survival conditions may also relate to conditions in which programmed cell death (apoptosis) pathways are activated or abrogated. A number of protein phosphatases are associated with the apoptosis pathways. Aberrations in the function of any one of the protein phosphatases could lead to cell immortality or premature cell death. [0220]
  • The term “aberration”, in conjunction with the function of a phosphatase in a signal transduction process, refers to a phosphatase that is over- or under-expressed in an organism, mutated such that its catalytic activity is lower or higher than wild-type protein phosphatase activity, mutated such that it can no longer interact with a natural binding partner, is no longer modified by another protein kinase or protein phosphatase, or no longer interacts with a natural binding partner. [0221]
  • The term “administering” relates to a method of incorporating a compound into cells or tissues of an organism. The abnormal condition can be prevented or treated when the cells or tissues of the organism exist within the organism or outside of the organism. Cells existing outside the organism can be maintained or grown in cell culture dishes. For cells harbored within the organism, many techniques exist in the art to administer compounds, including (but not limited to) oral, parenteral, dermal, injection, and aerosol applications. For cells outside of the organism, multiple techniques exist in the art to administer the compounds, including (but not limited to) cell microinjection techniques, transformation techniques and carrier techniques. [0222]
  • The abnormal condition can also be prevented or treated by administering a compound to a group of cells having an aberration in a signal transduction pathway to an organism. The effect of administering a compound on organism function can then be monitored. The organism is preferably a mouse, rat, rabbit, guinea pig or goat, more preferably a monkey or ape, and most preferably a human. [0223]
  • Stimulating or Antagonizing Phosphatase-Associated Activity [0224]
  • The present invention also encompasses a method of agonizing (stimulating) or antagonizing phosphatase associated activity in a mammal comprising administering to said mammal an agonist or antagonist to an amino acid sequence selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, a functional derivative thereof, and a fragment thereof in an amount sufficient to effect said agonism or antagonism. The present application also contemplates a method of treating diseases in a mammal with an agonist or antagonist of the activity of one of the above mentioned polypeptides of the invention comprising administering the agonist or antagonist to a mammal in an amount sufficient to agonize or antagonize a phosphatase-associated function. [0225]
  • The relevance of a phosphatase gene to a particular diseased condition can be evaluated in order to effect treatment. According to one embodiment of the present invention, microarray expression analysis is performed to establish expression profiles of various phosphatase genes according to the invention, and thereby identify the ones whose expression correlates with certain diseased conditions. [0226]
  • Due to the broad functional implications of various phosphatase families, such treatment may be effectuated to a wide range of diseases, including cancer, pathophysiological hypoxia, cardiovascular disorders, Papillon-Lefevre syndrome, Cowden disease, ectordermal dysplasia, Moebius syndrome, Bjornstad syndrome, Bannayan Zonana syndrome, schizophrenia and hamartomas. Of particular importance is treatment to various type of cancers. Accordingly, the present invention provides methods for treating pathologies, including breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer. [0227]
  • For example, cDNAs made from RNA samples of a variety of tissue sources were spotted onto nylon membranes and hybridized with radio-labeled probes derived from the phosphatase genes of interest. Referring to Example 3 and table 5, phosphatase gene sequences used include: SEQ ID NO4, SEQ ID NO: 5, and SEQ ID NO: 7. As discussed in the description of Table 5, infra, samples from normal tissues, tumor tissues, various cell lines, and P53 wild type and mutant were used to make the expression array. As shown in Example 3, the relative gene expression levels of the tested phosphatase genes in various tissue sources and cell lines were quantitated by measuring Syber Green I staining of hybridized signals. The numerical readings recorded in the table were normalized to the hybridization result from ds cDNA or undenatured probes, after subtracting the background counts. [0228]
  • Together with the information of corresponding nucleic acid and amino acid sequences provided herein, the relevant expression levels in Table 5 constitutes expression profiles of the phosphatase genes of interest in various tissue sources. Such expression profile data guides application of the treatment regime according to the present invention. For example, referring to the sample, “M14” cell line (a malignant melanoma) in Table 5, the levels of expression of SEQ ID NO: 4 is zero. The level of expression of SEQ ID NO: 7 (58) is low to marginal. However, the level of expression of SEQ ID NO: 5 (2,528) is significantly higher. Such horizontal comparison reveals that the phosphatase gene encoded by SEQ ID NO: 5 is implicated in melanoma. That is, manipulation of the function activities of this gene may affect the cancerous condition of malignant melanoma. SEQ ID NO: 5 (SGP003) encodes SEQ ID NO: 17, a protein belonging to the UP family, as shown in Table 1, for example. Therefore, a method of treating the cancer condition connected to a malignant melanoma can be, for example, to administer to the patient suffering from this cancer an agent that is capable of modulating the activities of the phosphatase activity of the protein represented by SEQ ID NO: 17. The expression analysis according to the preferred embodiment of this invention, thus, confers specificity and effectiveness to the method of treatment disclosed. [0229]
  • It should be appreciated that many ways of comparison and correlation analysis may be carried out, based on expression data generated in the way similar to that described in Example 3. These ways will be apparent to one skilled in the art, based on the above discussion and, therefore, fall within the scope of the invention. Inferences derived from those comparison and correlation analysis similarly may be used in substantiating a treatment method or regimen, according to the invention. For instance, when pairs of samples of normal tissues and diseased tissues are used to make the expression arrays, the data generated will specifically demonstrate which phosphatase genes are differentially expressed in certain diseased conditions and, thereby, form targets of the treatment method according to the present invention. That is, modulators or agents that are capable of regulating their activities, either in vivo or in vitro, may be identified and used in the treatment of the given diseased conditions. [0230]
  • According to the present invention, there also is provided a method for detecting a phosphatase in a sample as a diagnostic tool for a disease or disorder using nucleotide probes derived from the phosphatase gene sequences disclosed in the present invention, such as those disclosed herein. Due to the broad functional implications of various phosphatase families, such diagnostic measures may be used for a wide range of diseases, including cancer, pathophysiological hypoxia, cardiovascular disorders, Papillon-Lefevre syndrome, Cowden disease, ectordermal dysplasia, Moebius syndrome, Bjomstad syndrome, Bannayan Zonana syndrome, schizophrenia and hamartomas. Of particular importance is diagnose of various type of cancers. The diagnostic method of the present invention may be used to test for breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer. [0231]
  • In a similar vein, it is useful to determine the level of relevance of a phosphatase gene to a particular diseased condition in order to effect accurate diagnoses. Such determinations can be accomplished by performing microarray expression analysis according to one embodiment of this invention. The phosphatase genes whose expression correlates with certain diseased conditions may be identified by the procedure described above. [0232]
  • The data obtained from the microarray data also can be used to diagnose a patient who may be suffering from a particular pathology. A method of diagnosing the cancer condition connected to melanoma, according to the present invention is, therefore, to contact a test sample, which may be collected from a patient, with a nucleotide probe which is capable of hybridizing to the nucleic acid sequence which encodes the protein represented by SEQ ID NO: 17; and then to detect the presence of the hybridized probe:target pairs and to quantify the level of such hybridization as an indication of the cancer condition connected to neuroblastoma. The expression analysis according to the preferred embodiment of this invention, thus, confers specificity and effectiveness to the diagnostic method disclosed. [0233]
  • As discussed above, many ways of comparison and correlation analysis may be carried out based on expression data generated in the way similar to that described here; they also necessarily fall in the scope of the present invention. Inferences derived from those comparison and correlation analysis may similarly be used in substantiating the diagnostic method according to this invention. One scenario to be noted is when pairs of samples of normal tissues and diseased tissues are used to make the expression arrays, the data generated will specifically demonstrate which phosphatase genes are differentially expressed in certain diseased conditions, therefore may serve as diagnostic markers used in the aforementioned diagnostic method. [0234]
  • According to the present invention, there also is provided another method for detection of a phosphatase in a sample as a diagnostic tool for a disease or disorder by comparing a nucleic acid target region of the phosphatase genes disclosed in the present invention, such genes encoding the amino acid sequences listed in FIG. 2, with a control region; and then detecting differences in sequence or amount between the target region and control region as an indication of the disease or disorder. This method also may be used for diagnosing a wide range of diseases, including cancer, pathophysiological hypoxia, cardiovascular disorders, Papillon-Lefevre syndrome, Cowden disease, ectordermal dysplasia, Moebius syndrome, Bjomstad syndrome, Bannayan Zonana syndrome, schizophrenia and hamartomas. Of particular importance is diagnosis of various type of cancers. As the aforementioned diagnostic method, this particular method may similarly be used to test for breast cancer, urogenital cancer, prostate cancer, head and neck cancer, lung cancer, synovial sarcomas, renal cell carcinoma, non-small cell lung cancer, hepatocellular carcinoma, pancreatic endocrine tumors, stomach cancer, gliobastoma, colorectal cancer, and thyroid cancer. [0235]
  • A target region can be any particular region of interest in a phosphatase gene, such as an upstream regulatory region. Variations of sequence in an upstream regulatory region in a family of phosphatase often have functional implications some of which may be significant in bringing about certain diseased conditions. Changes of the amount of a target region, e.g., changes of number of copies of a regulatory region such as a receptor-binding site, in certain phosphatase genes, may also represent mechanisms of functional differentiation and hence may be connected to certain diseased states. Detection of such differences in sequence and amount of a target region compared to a control region therefore may effectively lead to detection of a diseased condition. [0236]
  • In one embodiment of the present invention, microarray studies may be used to identify the potential connections between a diseased condition and variations of a target region among a set of phosphatase genes. For example, nucleic acid probes may be made that correspond to a given target region and a control region, respectively, of a phosphatase gene of interest. Samples from normal and diseased tissues are used to make microarray as discussed, supra, and in Example 3. Hybridization of these probes to the array so made will yield comparative profiles of the region of interest in the normal and diseased condition, and thus may derive a definition of differences of the target region and control region that is characterized of the disease in question. Such definition, in turn, may serve as an indication of the diseased condition as used in the second-mentioned diagnostic method according to the present invention. It should be appreciated that many equivalent or similar methods may be used in carrying out the diagnosis according to this method which would become apparent to the skilled person in the art based on the example provided here, and therefore, they are covered in the scope of this invention. [0237]
  • In an effort to discover novel treatments for diseases, biomedical researchers and chemists have designed, synthesized, and tested molecules that inhibit the function of protein phosphatases. Some small organic molecules form a class of compounds that modulate the function of protein phosphatases. Examples of molecules that have been reported to inhibit the function of protein phosphatases include, but are not limited to, bis monocyclic, bicyclic or heterocyclic aryl compounds (PCT WO 92/20642, published Nov. 26, 1992 by Maguire et al.), vinylene-azaindole derivatives (PCT WO 94/14808, published Jul. 7, 1994 by Ballinari et al.), 1-cyclopropyl-4-pyridyl-quinolones (U.S. Pat. No. 5,330,992), styryl compounds (U.S. Pat. No. 5,217,999), styryl-substituted pyridyl compounds (U.S. Pat. No. 5,302,606), certain quinazoline derivatives (EP Application No. 0 566 266 A1), seleoindoles and selenides (PCT WO 94/03427, published Feb. 17, 1994 by Denny et al.), tricyclic polyhydroxylic compounds (PCT WO 92/21660, published Dec. 10, 1992 by Dow), and benzylphosphonic acid compounds (PCT WO 91/15495, published Oct. 17, 1991 by Dow et al). [0238]
  • Compounds that can traverse cell membranes and are resistant to acid hydrolysis are potentially advantageous as therapeutics as they can become highly bioavailable after being administered orally to patients. However, many of these protein phosphatase inhibitors only weakly inhibit the function of protein phosphatases. In addition, many inhibit a variety of protein phosphatases and will therefore cause multiple side-effects as therapeutics for diseases. [0239]
  • Some indolinone compounds, however, form classes of acid resistant and membrane permeable organic molecules. WO 96/22976 (published Aug. 1, 1996 by Ballinari et al.) describes hydrosoluble indolinone compounds that harbor tetralin, naphthalene, quinoline, and indole substituents fused to the oxindole ring. These bicyclic substituents are in turn substituted with polar moieties including hydroxylated alkyl, phosphate, and ether moieties. U.S. patent application Ser. No. 08/702,232, filed Aug. 23, 1996, entitled “Indolinone Combinatorial Libraries and Related Products and Methods for the Treatment of Disease” by Tang et al. (U.S. Ser. No. 08/702,232) and U.S. Pat. No. 5,880,141, entitled “Benzylidene-Z-Indoline Compounds for the Treatment of Disease” by Tang et al. (U.S. Ser. No. 08/485,323) and International Patent Publications WO 96/40116, published Dec. 19, 1996 by Tang, et al., and WO 96/22976, published Aug. 1, 1996 by Ballinari et al., all of which are incorporated herein by reference in their entirety, including any drawings, figures, or tables, describe indolinone chemical libraries of indolinone compounds harboring other bicyclic moieties as well as monocyclic moieties fused to the oxindole ring. Application Ser. No. 08/702,232, filed Aug. 23, 1996, entitled “Indolinone Combinatorial Libraries and Related Products and Methods for the Treatment of Disease” by Tang et al.; U.S. Pat. No. 5,880,141, filed Jun. 7, 1995, entitled “Benzylidene-Z-Indoline Compounds for the Treatment of Disease” by Tang et al. (U.S. Ser. No. 08/485,323), and WO 96/22976, published Aug. 1, 1996 by Ballinari et al. teach methods of indolinone synthesis, methods of testing the biological activity of indolinone compounds in cells, and inhibition patterns of indolinone derivatives. [0240]
  • Other examples of substances capable of modulating phosphatase activity include, but are not limited to, tyrphostins, quinazolines, quinoxolines, and quinolines. The quinazolines, tyrphostins, quinolines, and quinoxolines referred to above include well known compounds such as those described in the literature. For example, representative publications describing quinazolines include Barker et al., EPO Publication No. 0 520 722 Al; Jones et al., U.S. Pat. No. 4,447,608; Kabbe et al., U.S. Pat. No. 4,757,072; Kaul and Vougioukas, U.S. Pat. No. 5,316,553; Kreighbaum and Comer, U.S. Pat. No. 4,343,940; Pegg and Wardleworth, EPO Publication No. 0 562 734 Al; Barker et al., (1991) Proc. of Am. Assoc. for Cancer Research 32:327; Bertino, J. R., (1979) Cancer Research 3:293-304; Bertino, J. R., (1979) Cancer Research 9(2 part 1):293-304; Curtin et al., (1986) Br. J. Cancer 53:361-368; Fernandes et al., (1983) Cancer Research 43:1117-1123 ; Ferris et al. J. Org. Chem. 44(2):173-178; Fry et al., (1994) [0241] Science 265;1093-1095; Jackman et al., (1981) Cancer Research 51:5579-5586; Jones et al. J. Med. Chem. 29(6):1114-1118; Lee and Skibo, (1987) Biochemistry 26(23):7355-7362; Lemus et al., (1989) J. Org. Chem. 54:3511-3518; Ley and Seng, (1975) Synthesis 1975:415-522; Maxwell et al., (1991) Magnetic Resonance in Medicine 17:189-196 ; Mini et al., (1985) Cancer Research 45:325-330; Phillips and Castle, J. (1980) Heterocyclic Chem. 17(19):1489-1596; Reece et al., (1977) Cancer Research 47(11):2996-2999; Sculier et al., (1986) Cancer Immunol. and Immunother. 23, A65; Sikora et al., (1984) Cancer Letters 23:289-295; Sikora et al., (1988) Analytical Biochem. 172:344-355; all of which are incorporated herein by reference in their entirety, including any drawings.
  • Quinoxaline is described in Kaul and Vougioukas, U.S. Pat. No. 5,316,553, incorporated herein by reference in its entirety, including any drawings. [0242]
  • Quinolines are described in Dolle et al., (1994) J. Med. Chem. 37:2627-2629; MaGuire, J. (1994) Med. Chem. 37:2129-2131; Burke et al., (1993) J. Med. Chem. 36:425-432; and Burke et al. (1992) BioOrganic Med. Chem. Letters 2:1771-1774, all of which are incorporated by reference in their entirety, including any drawings. [0243]
  • Tyrphostins are described in Allen et al., (1993) Clin. Exp. Immunol. 91:141-156; Anafi et al., (1993) Blood 82:12, 3524-3529; Balker et al., (1992) J. Cell Sci. 102:543-555; Bilder et al., (1991) Amer. Physiol. Soc. pp. 6363-6143:C721-C730; Brunton et al., (1992) Proceedings of Amer. Assoc. Cancer Rsch. 33:558; Bryckaert et al., (1992) Exp. Cell Research 199:255-261; Dong et al., (1993) J. Leukocyte Biology 53:53-60; Dong et al., (1993) J. Immunol. 151(5):2717-2724; Gazit et al., (1989) J. Med. Chem. 32, 2344-2352; Gazit et al., (1993) J. Med. Chem. 36:3556-3564; Kaur et al., (1994) Anti-Cancer Drugs 5:213-222; King et al., (1991) Biochem. J. 275:413-418; Kuo et al., (1993) Cancer Letters 74:197-202; Levitzld, A., (1992) The FASEB J. 6:3275-3282; Lyall et al., (1989) J. Biol. Chem. 264:14503-14509; Peterson et al., (1993) The Prostate 22:335-345; Pillemer et al., (1992) Int. J. Cancer 50:80-85; Posner et al., (1993) Molecular Pharmacology 45:673-683; Rendu et al., (1992) Biol. Pharmacology 44(5):881-888; Sauro and Thomas, (1993) Life Sciences 53:371-376; Sauro and Thomas, (1993) J. Pharm. and Experimental Therapeutics 267(3):119-1125; Wolbring et al., (1994) J. Biol. Chem. 269(36):22470-22472; and Yoneda et al., (1991) Cancer Research 51:4430-4435; all of which are incorporated herein by reference in their entirety, including any drawings. [0244]
  • Other compounds that could be used as modulators include oxindolinones such as those described in U.S. patent application Ser. No. 08/702,232 filed Aug. 23, 1996, incorporated herein by reference in its entirety, including any drawings. [0245]
  • Recombinant DNA Technology: [0246]
  • DNA Constructs Comprising a Phosphatase Nucleic Acid Molecule and Cells Containing These Constructs. [0247]
  • The present invention also relates to a recombinant DNA molecule comprising, 5′ to 3′, a promoter effective to initiate transcription in a host cell and the above-described nucleic acid molecules. In addition, the present invention relates to a recombinant DNA molecule comprising a vector and an above-described nucleic acid molecule. The present invention also relates to a nucleic acid molecule comprising a transcriptional region functional in a cell, a sequence complementary to an RNA sequence encoding an amino acid sequence corresponding to the above-described polypeptide, and a transcriptional termination region functional in said cell. The above-described molecules may be isolated and/or purified DNA molecules. [0248]
  • The present invention also relates to a cell or organism that contains an above-described nucleic acid molecule and thereby is capable of expressing a polypeptide. The polypeptide may be purified from cells which have been altered to express the polypeptide. A cell is said to be “altered to express a desired polypeptide” when the cell, through genetic manipulation, is made to produce a protein which it normally does not produce or which the cell normally produces at lower levels. One skilled in the art can readily adapt procedures for introducing and expressing either genomic, cDNA, or synthetic sequences into either eukaryotic or prokaryotic cells. [0249]
  • A nucleic acid molecule, such as DNA, is said to be “capable of expressing” a polypeptide if it contains nucleotide sequences which contain transcriptional and translational regulatory information and such sequences are “operably linked” to nucleotide sequences which encode the polypeptide. An operable linkage is a linkage in which the regulatory DNA sequences and the DNA sequence sought to be expressed are connected in such a way as to permit gene sequence expression. The precise nature of the regulatory regions needed for gene sequence expression may vary from organism to organism, but shall in general include a promoter region which, in prokaryotes, contains both the promoter (which directs the initiation of RNA transcription) as well as the DNA sequences which, when transcribed into RNA, will signal synthesis initiation. Such regions will normally include those 5′-non-coding sequences involved with initiation of transcription and translation, such as the TATA box, capping sequence, CAAT sequence, and the like. [0250]
  • If desired, the [0251] non-coding region 3′ to the sequence encoding a phosphatase of the invention may be obtained by the above-described methods. This region may be retained for its transcriptional termination regulatory sequences, such as termination and polyadenylation. Thus, by retaining the 3′-region naturally contiguous to the DNA sequence encoding a phosphatase of the invention, the transcriptional termination signals may be provided. Where the transcriptional termination signals are not satisfactorily functional in the expression host cell, then a 3′ region functional in the host cell may be substituted.
  • Two DNA sequences (such as a promoter region sequence and a sequence encoding a phosphatase of the invention) are said to be operably linked if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the promoter region sequence to direct the transcription of a gene sequence encoding a phosphatase of the invention, or (3) interfere with the ability of the gene sequence of a phosphatase of the invention to be transcribed by the promoter region sequence. Thus, a promoter region would be operably linked to a DNA sequence if the promoter were capable of effecting transcription of that DNA sequence. Thus, to express a gene encoding a phosphatase of the invention, transcriptional and translational signals recognized by an appropriate host are necessary. [0252]
  • The present invention encompasses the expression of a gene encoding a phosphatase of the invention (or a functional derivative thereof) in either prokaryotic or eukaryotic cells. Prokaryotic hosts are, generally, very efficient and convenient for the production of recombinant proteins and are, therefore, one type of preferred expression system for phosphatases of the invention. Prokaryotes most frequently are represented by various strains of [0253] E. coli. However, other microbial strains may also be used, including other bacterial strains.
  • In prokaryotic systems, plasmid vectors that contain replication sites and control sequences derived from a species compatible with the host may be used. Examples of suitable plasmid vectors may include pBR322, pUC118, pUC119 and the like; suitable phage or bacteriophage vectors may include λgt10, λgt11 and the like; and suitable virus vectors may include pMAM-neo, pKRC and the like. Preferably, the selected vector of the present invention has the capacity to replicate in the selected host cell. [0254]
  • Recognized prokaryotic hosts include bacteria such as [0255] E. coli, Bacillus, Streptomyces, Pseudomonas, Salmonella, Seiratia, and the like. However, under such conditions, the polypeptide will not be glycosylated. The prokaryotic host must be compatible with the replicon and control sequences in the expression plasmid.
  • To express a phosphatase of the invention (or a functional derivative thereof) in a prokaryotic cell, it is necessary to operably link the sequence encoding the phosphatase of the invention to a functional prokaryotic promoter. Such promoters may be either constitutive or, more preferably, regulatable (i.e., inducible or derepressible). Examples of constitutive promoters include the int promoter of bacteriophage λ, the bla promoter of the β-lactamase gene sequence of pBR322, and the cat promoter of the chloramphenicol acetyl transferase gene sequence of pPR325, and the like. Examples of inducible prokaryotic promoters include the major right and left promoters of bacteriophage λ (P[0256] L and PR), the tip, recA, λacZ, λacI, and gal promoters of E. coli, the α-amylase (Ulmanen et al., J. Bacteriol. 162:176-182, 1985) and the ζ-28-specific promoters of B. subtilis (Gilman et al., Gene Sequence 32:11-20, 1984), the promoters of the bacteriophages of Bacillus (Gryczan, In: The Molecular Biology of the Bacilli, Academic Press, Inc., NY, 1982), and Streptomyces promoters (Ward et al., Mol. Gen. Genet. 203:468-478, 1986). Prokaryotic promoters are reviewed by Glick (Ind. Microbiot. 1:277-282, 1987), Cenatiempo (Biochimie 68:505-516, 1986), and Gottesman (Ann. Rev. Genet. 18:415-442, 1984).
  • Proper expression in a prokaryotic cell also requires the presence of a ribosome-binding site upstream of the gene sequence-encoding sequence. Such ribosome-binding sites are disclosed, for example, by Gold et al. (Ann. Rev. Microbiol. 35:365-404, 1981). The selection of control sequences, expression vectors, transformation methods, and the like, are dependent on the type of host cell used to express the gene. As used herein, “cell”, “cell line”, and “cell culture” may be used interchangeably and all such designations include progeny. Thus, the words “transformants” or “transformed cells” include the primary subject cell and cultures derived therefrom, without regard to the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. However, as defined, mutant progeny have the same functionality as that of the originally transformed cell. [0257]
  • Host cells which may be used in the expression systems of the present invention are not strictly limited, provided that they are suitable for use in the expression of the phosphatase polypeptide of interest. Suitable hosts may often include eukaryotic cells. Preferred eukaryotic hosts include, for example, yeast, fungi, insect cells, mammalian cells either in vivo, or in tissue culture. Mammalian cells which may be useful as hosts include HeLa cells, cells of fibroblast origin such as VERO or CHO-K1, or cells of lymphoid origin and their derivatives. Preferred mammalian host cells include SP2/0 and J558L, as well as neuroblastoma cell lines such as IMR 332, which may provide better capacities for correct post-translational processing. [0258]
  • In addition, plant cells are also available as hosts, and control sequences compatible with plant cells are available, such as the cauliflower mosaic virus 35S and 19S, and nopaline synthase promoter and polyadenylation signal sequences. Another preferred host is an insect cell, for example the Drosophila larvae. Using insect cells as hosts, the Drosophila alcohol dehydrogenase promoter can be used (Rubin, [0259] Science 240:1453-1459, 1988). Alternatively, baculovirus vectors can be engineered to express large amounts of phosphatases of the invention in insect cells (Jasny, Science 238:1653, 1987; Miller et al., In: Genetic Engineering, Vol. 8, Plenum, Setlow et al., eds., pp. 277-297, 1986).
  • Any of a series of yeast expression systems can be utilized which incorporate promoter and termination elements from the actively expressed sequences coding for glycolytic enzymes that are produced in large quantities when yeast are grown in mediums rich in glucose. Known glycolytic gene sequences can also provide very efficient transcriptional control signals. Yeast provides substantial advantages in that it can also carry outpost-translational modifications. A number of recombinant DNA strategies exist utilizing strong promoter sequences and high copy number plasmids which can be utilized for production of the desired proteins in yeast. Yeast recognizes leader sequences on cloned mammalian genes and secretes peptides bearing leader sequences (i.e., pre-peptides). Several possible vector systems are available for the expression of phosphatases of the invention in a mammalian host. [0260]
  • A wide variety of transcriptional and translational regulatory sequences may be employed, depending upon the nature of the host. The transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, cytomegalovirus, simian virus, or the like, where the regulatory signals are associated with a particular gene sequence which has a high level of expression. Alternatively, promoters from mammalian expression products, such as actin, collagen, myosin, and the like, may be employed. Transcriptional initiation regulatory signals may be selected which allow for repression or activation, so that expression of the gene sequences can be modulated. Of interest are regulatory signals which are temperature-sensitive so that by varying the temperature, expression can be repressed or initiated, or are subject to chemical (such as metabolite) regulation. [0261]
  • Expression of phosphatases of the invention in eukaryotic hosts requires the use of eukaryotic regulatory regions. Such regions will, in general, include a promoter region sufficient to direct the initiation of RNA synthesis. Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence Namer et al., J. Mol. Appl. Gen. 1:273-288, 1982); the TK promoter of Herpes virus (McKnight, [0262] Cell 31:355-365, 1982); the SV40 early promoter (Benoist et al., Nature (London) 290:304-31, 1981); and the yeast gal4 gene sequence promoter (Johnston et al., Proc. Natl. Acad. Sci. (USA) 79:6971-6975, 1982; Silver et al., Proc. Natl. Acad. Sci. (USA) 81:5951-5955, 1984).
  • Translation of eukaryotic mRNA is initiated at the codon which encodes the first methionine. For this reason, it is preferable to ensure that the linkage between a eukaryotic promoter and a DNA sequence which encodes a phosphatase of the invention (or a functional derivative thereof) does not contain any intervening codons which are capable of encoding a methionine (i.e., AUG). The presence of such codons results either in the formation of a fusion protein (if the AUG codon is in the same reading frame as the phosphatase of the invention coding sequence) or a frame-shift mutation (if the AUG codon is not in the same reading frame as the phosphatase of the invention coding sequence). [0263]
  • A nucleic acid molecule encoding a phosphatase of the invention and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a nonreplicating DNA or RNA molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule. Since such molecules are incapable of autonomous replication, the expression of the gene may occur through the transient expression of the introduced sequence. Alternatively, permanent expression may occur through the integration of the introduced DNA sequence into the host chromosome. [0264]
  • A vector may be employed which is capable of integrating the desired gene sequences into the host cell chromosome. Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the expression vector. The marker may provide for prototrophy to an auxotrophic host, biocide resistance, e.g., antibiotics, or heavy metals, such as copper, or the like. The selectable marker gene sequence can either be directly linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals. cDNA expression vectors incorporating such elements include those described by Okayama ([0265] Mol. Cell. Biol. 3:280-289, 1983).
  • The introduced nucleic acid molecule can be incorporated into a plasmid or viral vector capable of autonomous replication in the recipient host. Any of a wide variety of vectors may be employed for this purpose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies of the vector which are desired in a particular host; and whether it is desirable to be able to “shuttle” the vector between host cells of different species. [0266]
  • Preferred prokaryotic vectors include plasmids such as those capable of replication in [0267] E. coli (such as, for example, pBR322, ColEl, pSC101, pACYC 184, πVX; “Molecular Cloning: A Laboratory Manual”, 1989, supra). Bacillus plasmids include pC194, pC221, pT127, and the like (Gryczan, In: The Molecular Biology of the Bacilli, Academic Press, NY, pp. 307-329, 1982). Suitable Streptomyces plasmids include p1J101 (Kendall et al., J. Bacteriol. 169:4177-4183, 1987), and streptomyces bacteriophages such as φC31 (Chater et al., In: Sixth International Symposium on Actinomycetales Biology, Akademiai Kaido, Budapest, Hungary, pp. 45-54, 1986). Pseudomonas plasmids are reviewed by John et al. (Rev. Infect. Dis. 8:693-704, 1986), and Izaki (Jpn. J. Bacteriol. 33:729-742, 1978).
  • Preferred eukaryotic plasmids include, for example, BPV, vaccinia, SV40, 2-micron circle, and the like, or their derivatives. Such plasmids are well known in the art (Botstein et al., Miami Wntr. Symp. 19:265-274, 1982; Broach, In: “The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance”, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., p.445-470, 1981; Broach, [0268] Cell 28:203-204, 1982; Bollon et al., J. Clin. Hematol. Oncol. 10:39-48, 1980; Maniatis, In: Cell Biology: A Comprehensive Treatise, Vol. 3, Gene Sequence Expression, Academic Press, NY, pp. 563-608, 1980).
  • Once the vector or nucleic acid molecule containing the construct(s) has been prepared for expression, the DNA construct(s) may be introduced into an appropriate host cell by any of a variety of suitable means, i.e., transformation, transfection, conjugation, protoplast fusion, electroporation, particle gun technology, calcium phosphate-precipitation, direct microinjection, and the like. After the introduction of the vector, recipient cells are grown in a selective medium, which selects for the growth of vector-containing cells. Expression of the cloned gene(s) results in the production of a phosphatase of the invention, or fragments thereof. This can take place in the transformed cells as such, or following the induction of these cells to differentiate (for example, by administration of bromodeoxyuracil to neuroblastoma cells or the like). A variety of incubation conditions can be used to form the peptide of the present invention. The most preferred conditions are those which mimic physiological conditions. [0269]
  • Transgenic Animals: [0270]
  • A variety of methods are available for the production of transgenic animals associated with this invention. DNA can be injected into the pronucleus of a fertilized egg before fusion of the male and female pronuclei, or injected into the nucleus of an embryonic cell (e.g., the nucleus of a two-cell embryo) following the initiation of cell division (Brinster et al., [0271] Proc. Nat. Acad. Sci. USA 82:4438-4442, 1985). Embryos can be infected with viruses, especially retroviruses, modified to carry inorganic-ion receptor nucleotide sequences of the invention.
  • Pluripotent stem cells derived from the inner cell mass of the embryo and stabilized in culture can be manipulated in culture to incorporate nucleotide sequences of the invention. A transgenic animal can be produced from such cells through implantation into a blastocyst that is implanted into a foster mother and allowed to come to term. Animals suitable for transgenic experiments can be obtained from standard commercial sources such as Charles River (Wilmington, Mass.), Taconic (Germantown, N.Y.), Harlan Sprague Dawley (Indianapolis, Ind.), etc. [0272]
  • The procedures for manipulation of the rodent embryo and for microinjection of DNA into the pronucleus of the zygote are well known to those of ordinary skill in the art (Hogan et al., supra). Microinjection procedures for fish, amphibian eggs and birds are detailed in Houdebine and Chourrout ([0273] Experieiztia 47:897-905, 1991). Other procedures for introduction of DNA into tissues of animals are described in U.S. Pat. No. 4,945,050 (Sanford et al., Jul. 30, 1990).
  • By way of example only, to prepare a transgenic mouse, female mice are induced to superovulate. Females are placed with males, and the mated females are sacrificed by CO[0274] 2 asphyxiation or cervical dislocation and embryos are recovered from excised oviducts. Surrounding cumulus cells are removed. Pronuclear embryos are then washed and stored until the time of injection. Randomly cycling adult female mice are paired with vasectomized males. Recipient females are mated at the same time as donor females. Embryos then are transferred surgically. The procedure for generating transgenic rats is similar to that of mice (Hammer et al., Cell 63:1099-1112, 1990).
  • Methods for the culturing of embryonic stem (ES) cells and the subsequent production of transgenic animals by the introduction of DNA into ES cells using methods such as electroporation, calcium phosphate/DNA precipitation and direct injection also are well known to those of ordinary skill in the art (Teratocarcinomas and Embryonic Stem Cells, A Practical Approach, E. J. Robertson, ed., IRL Press, 1987). [0275]
  • In cases involving random gene integration, a clone containing the sequence(s) of the invention is co-transfected with a gene encoding resistance. Alternatively, the gene encoding neomycin resistance is physically linked to the sequence(s) of the invention. Transfection and isolation of desired clones are carried out by any one of several methods well known to those of ordinary skill in the art (E. J. Robertson, supra). [0276]
  • DNA molecules introduced into ES cells can also be integrated into the chromosome through the process of homologous recombination (Capecchi, [0277] Science 244:1288-1292, 1989). Methods for positive selection of the recombination event (i.e., neo resistance) and dual positive-negative selection (i.e., neo resistance and gancyclovir resistance) and the subsequent identification of the desired clones by PCR have been described by Capecchi, supra and Joyner et al. (Nature 338:153-156, 1989), the teachings of which are incorporated herein in their entirety including any drawings. The final phase of the procedure is to inject targeted ES cells into blastocysts and to transfer the blastocysts into pseudopregnant females. The resulting chimeric animals are bred and the offspring are analyzed by Southern blotting to identify individuals that carry the transgene. Procedures for the production of non-rodent mammals and other animals have been discussed by others (Houdebine and Chourrout, supra; Pursel et al., Science 244:1281-1288, 1989; and Simms et al., Bio/Technology 6:179-183, 1988).
  • Thus, the invention provides transgenic, nonhuman mammals containing a transgene encoding a kinase of the invention or a gene affecting the expression of the kinase. Such transgenic nonhuman mammals are particularly useful as an in vivo test system for studying the effects of introduction of a kinase, or regulating the expression of a kinase (i.e., through the introduction of additional genes, antisense nucleic acids, or ribozymes). [0278]
  • A “transgenic animal” is an animal having cells that contain DNA which has been artificially inserted into a cell, which DNA becomes part of the genome of the animal which develops from that cell. Preferred transgenic animals are primates, mice, rats, cows, pigs, horses, goats, sheep, dogs and cats. The transgenic DNA may encode human kinases. Native expression in an animal may be reduced by providing an amount of antisense RNA or DNA effective to reduce expression of the receptor. [0279]
  • Gene Therapy [0280]
  • Phosphatases or their genetic sequences will also be useful in gene therapy (reviewed in Miller, [0281] Nature 357:455-460, 1992). Miller states that advances have resulted in practical approaches to human gene therapy that have demonstrated positive initial results. The basic science of gene therapy is described in Mulligan (Science 260:926-931, 1993).
  • In one preferred embodiment, an expression vector containing a phosphatase coding sequence is inserted into cells, the cells are grown in vitro and then infused in large numbers into patients. In another preferred embodiment, a DNA segment containing a promoter of choice (for example a strong promoter) is transferred into cells containing an endogenous gene encoding phosphatases of the invention in such a manner that the promoter segment enhances expression of the endogenous phosphatase gene (for example, the promoter segment is transferred to the cell such that it becomes directly linked to the endogenous phosphatase gene). [0282]
  • The gene therapy may involve the use of an adenovirus containing phosphatase cDNA targeted to a tumor, systemic phosphatase increase by implantation of engineered cells, injection with phosphatase-encoding virus, or injection of naked phosphatase DNA into appropriate tissues. [0283]
  • Target cell populations may be modified by introducing altered forms of one or more components of the protein complexes in order to modulate the activity of such complexes. For example, by reducing or inhibiting a complex component activity within target cells, an abnormal signal transduction event(s) leading to a condition may be decreased, inhibited, or reversed. Deletion or missense mutants of a component, that retain the ability to interact with other components of the protein complexes but cannot function in signal transduction, may be used to inhibit an abnormal, deleterious signal transduction event. [0284]
  • Expression vectors derived from viruses such as retroviruses, vaccinia virus, adenovirus, adeno-associated virus, herpes viruses, several RNA viruses, or bovine papilloma virus, may be used for delivery of nucleotide sequences (e.g., cDNA) encoding recombinant phosphatase of the invention protein into the targeted cell population (e.g., tumor cells). Methods which are well known to those skilled in the art can be used to construct recombinant viral vectors containing coding sequences (Maniatis et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989; Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, N.Y., 1989). Alternatively, recombinant nucleic acid molecules encoding protein sequences can be used as naked DNA or in a reconstituted system e.g., liposomes or other lipid systems for delivery to target cells (e.g., Felgner et al., [0285] Nature 337:387-8, 1989). Several other methods for the direct transfer of plasmid DNA into cells exist for use in human gene therapy and involve targeting the DNA to receptors on cells by complexing the plasmid DNA to proteins (Miller, supra).
  • In its simplest form, gene transfer can be performed by simply injecting minute amounts of DNA into the nucleus of a cell, through a process of microinjection (Capecchi, [0286] Cell 22:479-88, 1980). Once recombinant genes are introduced into a cell, they can be recognized by the cell's normal mechanisms for transcription and translation, and a gene product will be expressed. Other methods have also been attempted for introducing DNA into larger numbers of cells. These methods include: transfection, wherein DNA is precipitated with calcium phosphate and taken into cells by pinocytosis (Chen et al., Mol. Cell Biol. 7:2745-52, 1987); electroporation, wherein cells are exposed to large voltage pulses to introduce holes into the membrane (Chu et al., Nucleic Acids Res. 15:1311-26, 1987); lipofection/liposome fusion, wherein DNA is packaged into lipophilic vesicles which fuse with a target cell (Felgner et al., Proc. Natl. Acad. Sci. USA. 84:7413-7417, 1987); and particle bombardment using DNA bound to small projectiles (Yang et al., Proc. Natl. Acad. Sci. 87:9568-9572, 1990). Another method for introducing DNA into cells is to couple the DNA to chemically modified proteins.
  • It has also been shown that adenovirus proteins are capable of destabilizing endosomes and enhancing the uptake of DNA into cells. The admixture of adenovirus to solutions containing DNA complexes, or the binding of DNA to polylysine covalently attached to adenovirus using protein crosslinking agents substantially improves the uptake and expression of the recombinant gene (Curiel et al., [0287] Am. J. Respir. Cell. Mol. Biol., 6:247-52, 1992).
  • As used herein “gene transfer” means the process of introducing a foreign nucleic acid molecule into a cell. Gene transfer is commonly performed to enable the expression of a particular product encoded by the gene. The product may include a protein, polypeptide, anti-sense DNA or RNA, or enzymatically active RNA. Gene transfer can be performed in cultured cells or by direct administration into animals. Generally gene transfer involves the process of nucleic acid contact with a target cell by non-specific or receptor mediated interactions, uptake of nucleic acid into the cell through the membrane or by endocytosis, and release of nucleic acid into the cytoplasm from the plasma membrane or endosome. Expression may require, in addition, movement of the nucleic acid into the nucleus of the cell and binding to appropriate nuclear factors for transcription. [0288]
  • As used herein “gene therapy” is a form of gene transfer and is included within the definition of gene transfer as used herein and specifically refers to gene transfer to express a therapeutic product from a cell in vivo or in vitro. Gene transfer can be performed ex vivo on cells which are then transplanted into a patient, or can be performed by direct administration of the nucleic acid or nucleic acid-protein complex into the patient. [0289]
  • In another preferred embodiment, a vector having nucleic acid sequences encoding a phosphatase polypeptide is provided in which the nucleic acid sequence is expressed only in specific tissue. Methods of achieving tissue-specific gene expression are set forth in International Publication No. WO 93/09236, filed Nov. 3, 1992 and published May 13, 1993. [0290]
  • In all of the preceding vectors set forth above, a further aspect of the invention is that the nucleic acid sequence contained in the vector may include additions, deletions or modifications to some or all of the sequence of the nucleic acid, as defined above. [0291]
  • In another preferred embodiment, a method of gene replacement is set forth. “Gene replacement” as used herein means supplying a nucleic acid sequence which is capable of being expressed in vivo in an animal and thereby providing or augmenting the function of an endogenous gene which is missing or defective in the animal. [0292]
  • Pharmaceutical Formulations and routes of Administration [0293]
  • The compounds described herein can be administered to a human patient per se, or in pharmaceutical compositions where it is mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. [0294]
  • Routes of Administration: [0295]
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections. [0296]
  • Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into a solid tumor, often in a depot or sustained release formulation. [0297]
  • Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with tumor-specific antibody. The liposomes will be targeted to and taken up selectively by the tumor. [0298]
  • Composition/Formulation: [0299]
  • The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping or lyophilizing processes. [0300]
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. [0301]
  • For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0302]
  • For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Suitable carriers include excipients such as, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [0303]
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [0304]
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. [0305]
  • For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. [0306]
  • For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. [0307]
  • The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0308]
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. [0309]
  • Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. [0310]
  • The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa buffer or other glycerides. [0311]
  • In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. [0312]
  • A pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent system may be the VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD:D5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose. [0313]
  • Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed. [0314]
  • The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. [0315]
  • Many of the tyrosine or serine/threonine phosphatase modulating compounds of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms. [0316]
  • Suitable Dosage Regimens: [0317]
  • Pharmaceutical compositions suitable for use in the present invention include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. [0318]
  • Methods of determining the dosages of compounds to be administered to a patient and modes of administering compounds to an organism are disclosed in U.S. application Ser. No. 08/702,282, filed Aug. 23, 1996 and International patent publication number WO 96/22976, published Aug. 1, 1996, both of which are incorporated herein by reference in their entirety, including any drawings, figures or tables. Those skilled in the art will appreciate that such descriptions are applicable to the present invention and can be easily adapted to it. [0319]
  • The proper dosage depends on various factors such as the type of disease being treated, the particular composition being used and the size and physiological condition of the patient. Therapeutically effective doses for the compounds described herein can be estimated initially from cell culture and animal models. For example, a dose can be formulated in animal models to achieve a circulating concentration range that initially takes into account the IC[0320] 50 as determined in cell culture assays. The animal model data can be used to more accurately determine useful doses in humans.
  • For any compound used in the methods of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC[0321] 50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the tyrosine or serine/threonine phosphatase activity). Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD[0322] 50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p.1).
  • Toxicity studies can also be carried out by measuring the blood cell composition. For example, toxicity studies can be carried out in a suitable animal model as follows: 1) the compound is administered to mice (an untreated control mouse should also be used); 2) blood samples are periodically obtained via the tail vein from one mouse in each treatment group; and 3) the samples are analyzed for red and white blood cell counts, blood cell composition and the percent of lymphocytes versus polymorphonuclear cells. A comparison of results for each dosing regime with the controls indicates if toxicity is present. [0323]
  • At the termination of each toxicity study, further studies can be carried out by sacrificing the animals preferably, in accordance with the American Veterinary Medical Association guidelines Report of the American Veterinary Medical Assoc. Panel on Euthanasia:229-249, 1993). Representative animals from each treatment group can then be examined by gross necropsy for immediate evidence of metastasis, unusual illness or toxicity. Gross abnormalities in tissue are noted and tissues are examined histologically. Compounds causing a reduction in body weight or blood components are less preferred, as are compounds having an adverse effect on major organs. In general, the greater the adverse effect the less preferred the compound. [0324]
  • For the treatment of cancers the expected daily dose of a hydrophobic pharmaceutical agent is between 1 to 500 mg/day, preferably 1 to 250 mg/day, and most preferably 1 to 50 mg/day. Drugs can be delivered less frequently provided plasma levels of the active moiety are sufficient to maintain therapeutic effectiveness. [0325]
  • Plasma levels should reflect the potency of the drug. Generally, the more potent the compound the lower the plasma levels necessary to achieve efficacy. [0326]
  • Plasma half-life and biodistribution of the drug and metabolites in the plasma, tumors and major organs can also be determined to facilitate the selection of drugs most appropriate to inhibit a disorder. Such measurements can be carried out. For example, “PLC analysis can be performed on the plasma of animals treated with the drag and the location of radiolabeled compounds can be determined using detection methods such as X-ray, CAT scan and MRI. Compounds that show potent inhibitory activity in the screening assays, but have poor pharmacokinetic characteristics, can be optimized by altering the chemical structure and retesting. In this regard, compounds displaying good pharmacokinetic characteristics can be used as a model. [0327]
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the phosphatase modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90% inhibition of the phosphatase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, “PLC assays or bioassays can be used to determine plasma concentrations. [0328]
  • Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. [0329]
  • In cases of local administration or selective uptake, the effective local concentration of the drag may not be related to plasma concentration. [0330]
  • The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. [0331]
  • Packaging: [0332]
  • The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the polynucleotide for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of a tumor, inhibition of angiogenesis, treatment of fibrosis, diabetes, and the like. [0333]
  • Functional Derivatives [0334]
  • Also provided herein are functional derivatives of a polypeptide or nucleic acid of the invention. By “functional derivative” is meant a “chemical derivative,” “fragment,” or “variant,” of the polypeptide or nucleic acid of the invention, which terms are defined below. A functional derivative retains at least a portion of the function of the protein, for example reactivity with an antibody specific for the protein, enzymatic activity or binding activity mediated through noncatalytic domains, which permits its utility in accordance with the present invention. It is well known in the art that due to the degeneracy of the genetic code numerous different nucleic acid sequences can code for the same amino acid sequence. Equally, it is also well known in the art that conservative changes in amino acid can be made to arrive at a protein or polypeptide that retains the functionality of the original. In both cases, all permutations are intended to be covered by this disclosure. [0335]
  • Included within the scope of this invention are the functional equivalents of the herein-described isolated nucleic acid molecules. The degeneracy of the genetic code permits substitution of certain codons by other codons that specify the same amino acid and hence would give rise to the same protein. The nucleic acid sequence can vary substantially since, with the exception of methionine and tryptophan, the known amino acids can be coded for by more than one codon. Thus, portions or all of the genes of the invention could be synthesized to give a nucleic acid sequence significantly different from one selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12. The encoded amino acid sequence thereof would, however, be preserved. [0336]
  • In addition, the nucleic acid sequence may comprise a nucleotide sequence which results from the addition, deletion or substitution of at least one nucleotide to the 5′-end and/or the 3′-end of the nucleic acid formula selected from the group consisting of those set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, or a derivative thereof. Any nucleotide or polynucleotide may be used in this regard, provided that its addition, deletion or substitution does not alter the amino acid sequence of selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, which is encoded by the nucleotide sequence. For example, the present invention is intended to include any nucleic acid sequence resulting from the addition of ATG as an initiation codon at the 5′-end of the inventive nucleic acid sequence or its derivative, or from the addition of TTA, TAG or TGA as a termination codon at the 3′-end of the inventive nucleotide sequence or its derivative. Moreover, the nucleic acid molecule of the present invention may, as necessary, have restriction endonuclease recognition sites added to its 5′-end and/or 3′-end. [0337]
  • Such functional alterations of a given nucleic acid sequence afford an opportunity to promote secretion and/or processing of heterologous proteins encoded by foreign nucleic acid sequences fused thereto. All variations of the nucleotide sequence of the phosphatase genes of the invention and fragments thereof permitted by the genetic code are, therefore, included in this invention. [0338]
  • Further, it is possible to delete codons or to substitute one or more codons with codons other than degenerate codons to produce a structurally modified polypeptide, but one which has substantially the same utility or activity as the polypeptide produced by the unmodified nucleic acid molecule. As recognized in the art, the two polypeptides are functionally equivalent, as are the two nucleic acid molecules that give rise to their production, even though the differences between the nucleic acid molecules are not related to the degeneracy of the genetic code. [0339]
  • A “chemical derivative” of the complex contains additional chemical moieties not normally a part of the protein. Covalent modifications of the protein or peptides are included within the scope of this invention. Such modifications may be introduced into the molecule by reacting targeted amino acid residues of the peptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues, as described below. [0340]
  • Cysteinyl residues most commonly are reacted with alpha-haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives. Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p-chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, or chloro-7-nitrobenzo-2-oxa-1,3-diazole. [0341]
  • Histidyl residues are derivatized by reaction with diethylprocarbonate at pH 5.5-7.0 because this agent is relatively specific for the histidyl side chain. Para-bromophenacyl bromide also is useful; the reaction is preferably performed in 0.1 M sodium cacodylate at pH 6.0. [0342]
  • Lysinyl and amino terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect or reversing the charge of the lysinyl residues. Other suitable reagents for derivatizing primary amine containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate. [0343]
  • Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pK[0344] a of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine alpha-amino group.
  • Tyrosyl residues are well-known targets of modification for introduction of spectral labels by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizol and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively. [0345]
  • Carboxyl side groups (aspartyl or glutamyl) are selectively modified by reaction with carbodiimide (R′—N—C—N—R′) such as 1-cyclohexyl-3-(2-morpholinyl(4-ethyl)carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl)carbodiimide. Furthermore, aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions. [0346]
  • Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention. [0347]
  • Derivatization with bifunctional agents is useful, for example, for cross-linking the component peptides of the protein to each other or to other proteins in a complex to a water-insoluble support matrix or to other macromolecular carriers. Commonly used cross-linking agents include, for example, 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3′-dithiobis(succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane. Derivatizing agents such as methyl-3-[p-azidophenyl)dithiolpropioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light. Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization. [0348]
  • Other modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the alpha-amino groups of lysine, arginine, and histidine side chains (Creighton, T. E., Proteins: Structure and Molecular Properties, W. H. Freeman & Co., San Francisco, pp. 79-86 (1983)), acetylation of the N-terminal amine, and, in some instances, amidation of the C-terminal carboxyl groups. [0349]
  • Such derivatized moieties may improve the stability, solubility, absorption, biological half-life, and the like. The moieties may alternatively eliminate or attenuate any undesirable side effect of the protein complex and the like. Moieties capable of mediating such effects are disclosed, for example, in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Co., Easton, Pa. (1990). [0350]
  • The term “fragment” is used to indicate a polypeptide derived from the amino acid sequence of the proteins, of the complexes having a length less than the full-length polypeptide from which it has been derived. Such a fragment may, for example, be produced by proteolytic cleavage of the full-length protein. Preferably, the fragment is obtained recombinantly by appropriately modifying the DNA sequence encoding the proteins to delete one or more amino acids at one or more sites of the C-terminus, N-terminus, and/or within the native sequence. Fragments of a protein are useful for screening for substances that act to modulate signal transduction, as described herein. It is understood that such fragments may retain one or more characterizing portions of the native complex. Examples of such retained characteristics include: catalytic activity; substrate specificity; interaction with other molecules in the intact cell; regulatory functions; or binding with an antibody specific for the native complex, or an epitope thereof. [0351]
  • Another functional derivative intended to be within the scope of the present invention is a “variant” polypeptide which either lacks one or more amino acids or contains additional or substituted amino acids relative to the native polypeptide. The variant may be derived from a naturally occurring complex component by appropriately modifying the protein DNA coding sequence to add, remove, and/or to modify codons for one or more amino acids at one or more sites of the C-terminus, N-terminus, and/or within the native sequence. It is understood that such variants having added, substituted and/or additional amino acids retain one or more characterizing portions of the native protein, as described above. [0352]
  • A functional derivative of a protein with deleted, inserted and/or substituted amino acid residues may be prepared using standard techniques well-known to those of ordinary skill in the art. For example, the modified components of the functional derivatives may be produced using site-directed mutagenesis techniques (as exemplified by Adelman et al., 1983, DNA 2:183) wherein nucleotides in the DNA coding the sequence are modified such that a modified coding sequence is modified, and thereafter expressing this recombinant DNA in a prokaryotic or eukaryotic host cell, using techniques such as those described above. Alternatively, proteins with amino acid deletions, insertions and/or substitutions may be conveniently prepared by direct chemical synthesis, using methods well-known in the art. The functional derivatives of the proteins typically exhibit the same qualitative biological activity as the native proteins. [0353]
  • The invention also provides methods for determining whether a nucleic acid sequence encodes a phosphatase, according to the invention, which contains one or more characterizing portions of the native complex. As noted, examples of such retained characteristics include: catalytic activity; substrate specificity; interaction with other molecules in the intact cell; regulatory functions; or binding with an antibody specific for the native complex, or an epitope thereof. Accordingly, the invention provides an assay analyzing one or more characteristics—in particular, the presence of a catalytic domain—of a polypeptide phosphatase encoded by a given nucleic acid molecule. [0354]
  • To this end, a suitable assay can begin by purifying and quantitating a photphase protein. The protein then can be assayed, for example, by serial dilution and incubation in a buffer (e.g. ABT buffer) comprising a substrate capable of undergoing hydrolysis and optionally a reducing agent capable of increasing any catalytic activity of the polypeptide. Preferably, the substrate is p-nitrophenyl phosphate (pNPP) and the reducing agent is dithiothreitol (DTT), at mM concentrations of 4× and 1×, respectively. Incubation can be at room temperature from about 2 minutes to overnight, depending on activity. To stop the reaction, add NaOH, which can be about 100 ul of 10 N NaOH. The suspension can be centrifuged and the supernatant analyzed at an OD of 410 nM to determine whether to protein phosphatase exhibited catalytic properties. [0355]
  • Tables and Description Thereof [0356]
  • Table 1 documents the name of each gene, the classification of each gene product, the positions of the open reading frames within the sequence, and the length of the corresponding peptide. From left to right the data presented is as follows: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Superfamily”, “Group”, “Family”, “NA_length”, “OUF Start”, “ORF End”, “ORF Length”, and “AA_length”. “Gene name” refers to name given the sequence encoding the phosphatase or phosphatase-like enzyme. Each gene is represented by “SGP” designation followed by an arbitrary number. The SGP name usually represents multiple overlapping sequences built into a single contiguous sequence (a “contig”). The “ID#na” and “ID#aa” refer to the identification numbers given each nucleic acid and amino acid sequence in this patent. “FL/Cat” refers to the length of the gene, with FL indicating full length, and “Cat” indicating that only the catalytic domain is presented. “Partial” in this column indicates that the sequence encodes a partial protein phosphatase catalytic domain. “Superfamily” identifies whether the gene is a dual specificity phosphatase, a protein tyrosine phosphatase or a serine threonine phosphatase. “Group” and “Family” refer to the phosphatase classification defined by sequence homology and based on previously established phylogenetic ([0357] The Protein Phosphatase Factsbook, Nick Tonks, Shirish Shenolikar, Harry Charbonneau, Academic Pr, 2000). “NA_length” refers to the length in nucleotides of the corresponding nucleic acid sequence. “ORF start” refers to the beginning nucleotide of the open reading frame. “ORF end” refers to the last nucleotide of the open reading frame, including the stop codon. “ORF length” refers to the length in nucleotides of the open reading frame. “AA length” refers to the length in amino acids of the peptide encoded in the corresponding nuclei acid sequence.
    TABLE I
    Open Reading Frames
    424454_2
    Gene
    Name ID#na ID#aa FL/Cat Superfamily Group Family NA_length ORF Start ORF End ORF Length AA_length
    SGP006
    1 13 FL Dual Phosphatase DSP MKP 6374 34 3183 3150 1049
    SGP002 2 14 FL Dual Phosphatase DSP MKP 2732 538 2535 1998 665
    SGP001 3 15 FL Dual Phosphatase DSP MKP 2260 709 2205 1497 498
    SGP018 4 16 FL Dual Phosphatase DSP MKP 4361 208 3609 3402 1133
    SGP003 5 17 FL Dual Phosphatase DSP MKP 1262 240 902 663 220
    SGP014 6 18 FL Dual Phosphatase DSP MKP 1917 31 1680 1650 549
    SGP060 7 19 FL Dual Phosphatase DSP MKP 636 1 636 636 211
    SGP008 8 20 FL Dual Phosphatase DSP STYX 1326 1 990 990 329
    SGP039 9 21 FL Serine Phosphatase STP PP2C 1083 1 1083 1083 360
    SGP040 10 22 FL Serine Phosphatase STP PP2C 1725 1 1725 1725 574
    SGP012 11 23 Cat Tyrosine Phosphatase RPTP PTPd 4719 1 4719 4719 1573
    SGP024 12 24 Partial Tyrosine Phosphatase RPTP PTPd 354 1 357 357 118
  • Table 2 lists the following features of the genes described in this application: chromosomal localization, single nucleotide polymorphisms (SNPs), representation in dbEST, and repeat regions. From left to right the data presented is as follows: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Superfamily”, “Group”, “Family”, “Chromosome”, “SNPs”, “dbEST_hits”, & “Repeats”. The contents of the first 7 columns (i.e.,. “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Superfamily”, “Group”, “Family”) are as described above for Table 1. “Chromosome” refers to the cytogenetic localization of the gene. Information in the “SNPs” column describes the nucleic acid position and degenerate nature of candidate single nucleotide polymorphisms (SNPs. “dbEST hits” lists accession numbers of entries in the public database of ESTs (dbEST, http://www.ncbi.nlm.nih.gov/dbEST/index.html) that contain at least 100 bp of 100% identity to the corresponding gene. These ESTs were identified by blastn of dbEST. “Repeats” contains information about the location of short sequences, approximately 20 bp in length, that are of low complexity and that are present in several distinct genes. These repeats were identified by blastn of the DNA sequence against the non-redundant nucleic acid database at NCBI (nrna). To be included in this repeat column, the sequence typically has 100% identity over its length and is present in at least 5 different genes. [0358]
    TABLE 2
    CHR, SMPs, dbEST, Repeats
    424454_2
    Gene
    Name ID#na ID#aa FL/Cat Superfamily Group Family Chromosome SNPs dbEST_hits Repeats
    SGP006 1 13 FL Dual DSP MKP 12q21.3-q22 6222 = R BE793092.1, Alu 5750-6010;
    Phosphatase (ccaaacataagtggcacar) AI651213.1, 5750-5770;
    dbSNP[rs881179 BE256978.1
    SGP002 2 14 FL Dual DSP MKP 12p11.1-p12.1 none BE897795; 2610-2631
    Phosphatase
    SGP001 3 15 FL Dual DSP MKP Xp11.1-11.3 none AI272231, 579-598
    Phosphatase BF206586,
    SGP018 4 16 FL Dual DSP MKP NA 2929 = M BF114681 993-1014
    Phosphatase (agaagatgtctgagtacm)
    dbSMP[ss1765941;
    1161 = S
    (catctaccccaatgas)
    dbSNP]ss1755940
    SGP003 5 17 FL Dual DSP MKP CHR10 none none 311-334
    Phosphatase
    SGP014 6 18 FL Dual DSP MKP 10q21.3 none AA723271, none
    Phosphatase AW444890.1,
    AA435513.1
    SGP060 7 19 FL Dual DSP MKP 8p11.1-q11.1 none BF207232, none
    Phosphatase centromeric BF314818,
    AW953216.1
    SGP008 8 20 FL Dual DSP STYX 20q11.2 871 = S AW406620, 1251-1270
    Phosphatase (cagcagcctccgagggaaccs) BF377364.1,
    dbSNP[ss1389419 AW593296.1
    SGP039 9 21 FL Serine STP PP2C NA none BE147139.1 none
    Phosphatase
    SGP040 10 22 FL Serine STP PP2C 8q21.3 none AV706533.1, none
    Phosphatase AV705571.1,
    AV710801.1
    SGP012 11 23 Cat Tyrosine RPTP PTPd NA none AL042532.1, 3105-3124
    Phosphatase A1381571,
    AW672677
    SGP024 12 24 Partial Tyrosine RPTP PTPd NA none none none
    Phosphatase
  • Table 3 lists the extent and the boundaries of the phosphatase catalytic domains. The column headings are: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Domain”, “Phos_start”, “Phos_end”, “Profile_start”, “Profile_end”. The contents columns “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, are as described above for Table 1. “Phos_Start”, “Phos_End”, “Profile_Start” and “Profile_End” refer to data obtained using a Hidden-Markov Model to define catalytic range boundaries (http://pfam.wustl.edu/index.html). The boundaries of the catalytic domains within the overall protein are noted in the “Phos Start” and “Phos End” columns. Three profiles were used, one for dual specificity phosphatases (DSP) which is 173 amino acids long;, one for STPs, which is 301 amino acids long; and one for PTPs, which is 264 amino acids long. (The profiles used are described in http://pfam.wustl.edu/). Proteins in which the profile recognizes a full length catalytic domain have a “Profile Start” of 1 and, for the three families, the following Profile Ends: 173 for DSP, 301 for STPs, and 264 for PTPs. Genes which have a partial catalytic domain will have a “Profile Start” of greater than 1 (indicating that the beginning of the phosphatase domain is missing, and/or a “Profile End” of less than 261 (indicating that the C-terminal end of the phosphatase domain is missing). Each of the sequences encompasses a complete catalytic domain, except for SGP024, which has a partial catalytic domain represents amino acids 205 to 264 of the PTP profile. [0359]
    TABLE 3
    Phosphatase Domains
    424454_2
    Gene Name ID#na ID#aa FL/Cat Domain Phos_start Phos_end Profile_start Profile_end
    SGP006
    1 13 FL DSP 308 446 1 173
    SGP002 2 14 FL DSP 158 297 1 173
    SGP001 3 15 FL DSP 307 441 1 173
    SGP018 4 16 FL DSP 185 330 1 173
    SGP003 5 17 FL DSP 54 199 1 173
    SGP014 6 18 FL 2 DSPs (37-181 & 368-520) 37 & 368 181 & 520 1 173
    SGP060 7 19 FL DSP 61 204 1 173
    SGP008 8 20 FL DSP 98 235 1 173
    SGP039 9 21 FL Protein phosphatase 2C 91 344 1 301
    SGP040 10 22 FL Protein phosphatase 2C 209 497 1 301
    SGP012 11 23 Cat PTP 1010 1259 1 264
    SGP024 12 24 Partial PTP 3 58 205 264
  • Table 4 describes the results of Smith Waterman similarity searches (Matrix: Pam100; gap open/[0360] extension penalties 12/2) of the amino acid sequences against the NCBI database of non-redundant protein sequences (http://www.ncbi.nlm.nih.gov/Entrez/protein.html). The column headings are: “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, “Family”, “Pscore”, “aa_length”, “aa_ID_match”, “% Identity”, “% Similar”, “ACC#_nraa_match”, “Description”, “Query start”, “Query end”, “Target start”, and “Target end”. The contents of columns, “Gene Name”, “ID#na”, “ID#aa”, “FL/Cat”, and “Family” are as described above for Table 1. “Pscore” refers to the Smith Waterman probability score. This number approximates the probability that the alignment occurred by chance. Thus, a very low number, such as 2.10E-64, indicates that there is a very significant match between the query and the database target. “aa_length” refers to the length of the protein in amino acids. “aa_ID_match” indicates the number of amino acids that were identical in the alignment. “% Identity” lists the percent of nucleotides that were identical over the aligned region. “% Similarity” lists the percent of amino acids that were similar over the alignment. “ACC#nraa_match” lists the accession number of the most similar protein in the NCBI database of non-redundant proteins. “Description” contains the name of the most similar protein in the NCBI database of non-redundant proteins. “Query start” refers to the amino acid number in the phosphatase (“Query”) at which the alignment begins. “Query end” refers to the amino acid number in the phosphatase (“Query”) at which the alignment ends. “Target start” refers to the amino acid number in the Smith Waterman hit (“Target”) from NRAA at which the alignment begins. “Target end” refers to the amino acid number in the Smith Waterman hit (“Target”) from NRAA at which the alignment ends. Note that for SGP006 there three entries, and for SGP014 there are two entries. These additional rows describe different regions of alignments with different database “Targets” (see below for detailed descriptions).
    TABLE 4
    Smith Waterman
    424454_2
    Gene % %
    Name ID#na ID#aa FL/Cat Family Pscore aa_length aa_ID_match Identity Similar ACC#_nraa_match
    SGP006 1 13 FL MKP 0 1049 715 100 100 BAA92536.1
    SGP006 1 13 FL MKP 1.50E−99 1049 245 46 59 BAA89534.1
    SGP006 1 13 FL MKP 6.80E−58 1049 119 41 59 NP_060327.1
    SGP002 2 14 FL MKP 1.10E− 665 304 46 60 NP_004411.1
    157
    SGP001 3 15 FL MKP 8.30E− 496 250 47 60 BAA89534.1
    133
    SGP018 4 16 FL MKP 2.20E−27 1133 79 45 63 NP_057448.1
    SGP003 5 17 FL MKP 3.40E−54 220 91 49 66 NP_057446.1
    SGP014 6 18 FL MKP 7.50E− 549 195 58 58 NP_057446.1
    122
    SGP014 6 18 FL MKP 8.20E−36 549 75 45 65 NP_004081.1
    SGP060 7 19 FL MKP 1.10E−46 211 65 53 72 NP_057448.1
    SGP008 8 20 FL STYX 4.40E− 329 250 92 92 CAC10005.1
    172
    SGP039 9 21 FL PP2C 1.00E− 360 164 95 99 AAD17235.1
    106
    SGP040 10 22 FL PP2C 9 574 574 100 100 NP_060914.1
    SGP012 11 23 Cat TPd 0.00E+00 1573 1053 90 70 NP_031981.1
    SGP024 12 24 Partial PTPd 5.90E−54 118 90 76 52 CAA38068.1
    Gene Query Query Target Target
    Name Description start end start end
    SGP006 KIAA1298 protein [Homo sapiens] 322 1049 11 738
    SGP006 MAP kinase phosphatase [Drosophila melanogaster] 120 477 199 551
    SGP006 Hypothetical protein FLJ20515 [Homo sapiens] 1 263 1 263
    SGP002 dual specificity phosphatase 8 [Homo sapiens] 13 665 14 625
    SGP001 MKP [Drosophila melanogaster] 1 442 1 522
    SGP018 Protein phosphatase LOC51207 [Homo sapiens] 162 334 23 194
    SGP003 Protein phosphatase LOC51207 [Homo sapiens] 22 206 12 197
    SGP014 Protein phosphatase LOC51207 [Homo sapiens] 324 549 1 198
    SGP014 Dual specificity phosphatase 3 [Homo sapiens] 20 179 6 174
    SGP060 Protein phosphatase LOC51207 [Homo sapiens] 45 205 31 191
    SGP006 Novel protein [Homo sapiens] 63 329 1 275
    SGP039 PP 2C [Mus musculus] 133 299 1 167
    SGP040 Pyruvate dehydrogenase phosphatase [Homo sapiens] 1 574 1 574
    SGP012 Embryonic stem cell phosphatase [Mus musculus] 1 1559 1 1704
    SGP024 Protein-tyrosine phosphatase delta [Homo sapiens] 1 118 1165 1292
  • Table 5 shows the results of a gene expression analysis of selected phosphatases presented in this application using a microarray of cDNAs derived from 499 tissues and cell lines. The cDNAs were spotted on nylon and probed with labeled phosphatase genes, as described in Materials and Methods below. The phosphatase probes were PCR cloned from genomic exons. Data presentation from left to right is as follows: “ID”: number of the sample; “Sample name”; “T/N”, tumor or normal tissue; “Type”, tissue of origin; “Tissue/cell line”, sample is derived from tissue or from a cultured cell line; “Notes”: additional information about the sample; “Treatment”: chemical or physical treatment of the tissue or cell line; “p53” refers to the status, mutant or wild-type, of the p53 gene in the source samples. Normalized expression values are presented for each gene referred to by its SGP and SEQ_ID# on the subsequent columns. Genes represented in Table 5 are: SGP003, SGP060, and SGP018. [0361]
  • Images of the blots containing the probed tissue arrays are included. [0362]
    TABLE 5
    Tissue Array
    424454_2
    ID#NA-4, ID#NA_s, ID#NA_7,
    ID Sample_name T/N Type Tissue/cell line Notes Treatment p53 SGP018 SGP003 SGP080
    7 cerebellum-h N neuro tissue none 738 0 1,924
    195 458 medulla mRNA T neuro tissue none 717 2,956 0
    11 fetal kidney-h N renal tissue none 708 3,029 341
    41 Duodenum-h N col tissue none 633 0 0
    8 pituitary gland-h N neuro tissue none 627 2,928 3,003
    14 salivary gl.-h N salivary tissue none 608 2,044 193
    31 trachea-h N trachea tissue none 580 0 0
    44 testis-h N testes tissue none 535 0 457
    65 HT154 T HNS tissue none 496 1,852 109
    165 ACHN T renal cell line Renal adenocarcinomca none 495 1,114 0
    1 adrenal gland-h N adrenal tissue none 470 0 804
    10 placenta-h N plac tissue none 459 294 0
    26 HPABC N endo cell line renal proximal tubule epithelial cells none 458 133 226
    6 pancreas-h N pan tissue none 451 1,691 0
    377 OVCAR-5-7 T OV cell line 400 ng/ml noco-24 mutant 448 0 0
    17 heart-h N heart tissue none 447 288,691 426
    43 Salivary gl.-h N salivary tissue none 446 0 68
    13 fetal liver-h N liver tissue none 437 1,858 0
    46 h keratinocytes, Feb. 25, 1992 #10 T keratinocyte cell line unknown 433 434 155
    21 liver-h N liver tissue none 408 0 56
    23 lung-h N lung tissue none 406 1,349 0
    457 SF-268-0 T neuro cell line 10 μM cisplatin mutant 405 0 0
    38 fetal liver-h N liver tissue none 393 160 41
    350 HT29-4 T col cell line 3 mM HU mutant 393 0 0
    26 TCGP T tissue tissue none 384 831 0
    324 Malme-3M T renal cell line Malignant melanoma, metastasis to none 382 1,223 279
    lung
    114 SF-539 T neuro cell line Glioblastoma none 375 0 268
    229 HL-60 T col cell line PML Peripheral blood, none 358 0 0
    proaryelocytci leukemia
    30 RPTBC N endo cell line mammary epithelial cells none 352 5,306 0
    375 OVCAR-5-6 T OV cell line 10 μM cisplatin mutant 350 0 0
    465 AngloTest1-13 N HUVEC cell line 10 mm stimulation with PDGF PDGF 341 0 0
    93 HT385 T lung tissue none 339 370 151
    111 A549/ATCC T lung cell line Lung carcinoma none 336 2,559 190
    423 C33A-8 T cervical cell line 400 ng/ml noco-48 mutant 333 0 0
    45 458 medulla RNA T neuro none 333 0 348
    354 HT29-6 T col cell line 10 μM cisplatin mutant 328 0 0
    15 fetal lung-h N lung tissue none 323 4,701 0
    483 Prostate_sampleMG-6 unknown pro unknown unknown 321 0 0
    143 UO-31 T renal cell line none 319 0 5
    5 brain-h N neuro tissue none 316 0 1,102
    468 AngloTest1-3 T endo cell line HeLa25X DEF-MES for Hypoxia, 4b 25X DEF-MES 304 0 0
    96 OVCAR-8 T OV cell line none 302 340 0
    76 HT317 T lung tissue none 300 0 0
    256 MDA-N T breast cell line none 296 1,080 0
    83 HELA-9h-031899 T endo cell line 294 0 28
    223 HT382 T lung tissue none 293 0 282
    22 Spleen-h N home tissue none 291 3,580 0
    25 testis-h N testes tissue none 290 0 197
    50 HT213 T kidney tissue none 265 347 0
    123 RPMI 8226 T LEU cell line Multiple anyeloma none 282 0 106
    201 stomach-h N col tissue none 282 0 0
    490 Prostate_sampleMG-10 unknown neuro unknown NIH3T3 vector unknown 279 0 0
    251 HT382-normal N lung tissue none 279 0 80
    378 MCF-7-1 T breast cell line normal/10% FBS none wt 274 0 0
    489 OVCAR-5-5 T OV cell line 2 μM AUR2 Inhibitor mutant 273 0 0
    121 HL-60 T col cell line PML Peripheral blood, none 273 0 0
    prorayclocytic leukemia
    323 BioMarker_BS-13 N cado cell line HUVEC VEGF + 5416-24h VEGF 273 0 0
    265 KHOS poly A+ T bone cell line none 271 801 83
    318 A498 T renal cell line Kidney carcinoma none 268 221 0
    139 HCT-15 T col cell line Colon adenocarcinoma none 267 0 0
    426 U2OS-2 T bone cell line low serum/0.1% FBS low serum mutant 267 0 0
    105 NCI-H322M T lung cell line Lung Br, A./Lung none 267 44 58
    bronchioloaceolar carcinoma
    85 HT146 T kidney tissue none 266 1,305 0
    411 C33A-2 T cervical cell line low serum/0.1% FBS low serum mutant 265 0 0
    464 Angio Test1-1 T endo cell line HeLa25X DEF-MBS for Hypoxia, 0h 25X DEF-MBS 260 0 0
    469 Prostate_sampleMG-21 unknown pan unknown He294 unknown 258 0 0
    254 MDA-MB-435 T breast cell line none 254 0 153
    82 HT335 T pan tissue none 253 0 176
    459 SF-266-7 T neuro cell line 400 ng/ml noco-24 mutant 253 0 0
    106 SNB-75 T neuro cell line Astrocytoma none 251 655 40
    92 HBLA-10h-031899 T endo cell line 250 0 0
    37 Skeletal muscle-h N muscle tissue none 250 0 113
    266 HT394 T lung tissue none 242 0 0
    319 HT393 T lung tissue none 240 0 106
    277 HT338 T lung tissue none 239 82 0
    237 UO-31 T renal cell line none 238 0 0
    456 EKVX-4 T lung cell line 3 mM HU mutant 237 0 0
    382 MCF-7-3 T breast cell line 200 μM mimosine wt 236 0 0
    410 SW480-6 T col cell line 10 μM cisplatin mutant 234 0 0
    476 AngloTest1-7 N cado cell line HUVEC unstimulated/control none 234 0 0
    369 SF539-6 T neuro cell line 10 μM cisplatin wt 234 0 0
    379 ADR-RES-5 T breast cell line 2 μM AUR2 inhibitor mutant 232 0 0
    74 HT314 T MG tissue none 225 730 172
    151 MCF7 T breast cell line Breast adenocarcinoma, pural none 225 88 0
    effusion
    436 U2OS-7 T bone cell line 400 ng/ml noco-24 mutant 224 0 0
    227 MOLT-4 T LEU cell line ALL Peripheral blood, acute none 223 0 0
    lymphoblastic leukemia
    359 SF539-1 T neuro cell line normal/10% FBS none wt 222 0 0
    412 SW480-7 T col cell line 400 ng/ml noco-24 mutant 221 0 0
    304 HOP-62 T lung cell line Lung adenocarcinoma none 219 0 0
    29 thyroid gland-h N thyroid tissue none 217 383 0
    399 HeLa-8 T endo cell line 400 ng/ml noco-48 HPV E6 215 0 0
    148 NCI-H322M T lung cell line Lung Br. A./Lung none 215 0 38
    bronchiovascular carcinoma
    218 BioMarker_BS-6 N endo cell line HUVEC VEGF-6h VEGF 213 0 0
    4 mammary gland-h N breast tissue none 213 870 0
    305 MDA-MB-231 T breast cell line Breast adenocarcinoma, pleural none 210 403 0
    effusion
    148 NCI-1460 T lung cell line Lung large cell carcinoma none 210 0 297
    202 Heart-h N heart tissue h sheriocarcinoma none 206 0 0
    80 HT327 T lung tissue none 208 0 0
    34 HCABC N cado cell line none 207 884 14
    104 SNB-19 T neuro cell line Glioblastoma none 207 0 11
    266 HT392 T lung tissue none 206 412 366
    312 SW-630 T col cell line Colon adenocarcinoma, lymph node none 204 0 123
    metastasis
    491 Prostate_sampleMG-11 unknown neuro unknown NIH3T3 EWS/FLII unknown 202 0 0
    156 SF-268 T neuro cell line Glioblastoma none 202 0 157
    107 NCI-H460 T lung cell line Lung large cell carcinoma none 201 0 0
    101 NCI-H23 T lung cell line Lung adenocarcinoma none 201 0 0
    441 Wt 38-5 N lung cell line 2 μM AUR2 inhibitor wt 200 0 0
    360 MCF-7-2 T breast cell line low serum/0.1% FBS low serum wt 200 0 0
    67 HT169 T pro tissue none 200 0 0
    118 K-562 T LEU cell line CML Chronic myelogenous leukemia none 199 0 0
    403 H1299-6 T lung cell line 10 μM cisplatin mutant 199 0 0
    310 HCC-3998 T col cell line none 196 0 0
    433 Wt 38-1 N lung cell line normal/10% FBS none wt 196 0 0
    53 fetal lung-h N lung tissue none 196 0 0
    314 COLO 305 T col cell line Colon adenocarcinoma none 194 0 242
    122 SN12C T neuro cell line none 194 0 0
    453 SF-268-4 T neuro cell line 3 mM HU mutant 193 0 0
    20 spinal conf-h N neuro tissue none 193 13,667 387
    97 HOP-92 T lung cell line Lung large cell carcinoma none 192 0 365
    77 Medulloblastoma #425 1 1/8 T neuro tissue h Wilms' tumor none 190 0 138
    259 adrenal gland-h N adrenal tissue none 185 4,748 0
    257 Y79 poly A+ T retinal cell line h retinoblastoma none 185 0 0
    79 HELA-2h-031899 T endo cell line 184 82 191
    155 Hs 578T T breast cell line Ductal carcinoma none 183 1,362 142
    35 Pancreas-h N pan tissue h embryonic pelatal mesenchyme none 183 851 0
    55 heart-h N heart tissue none 182 2 0
    187 UACC-62 T mel cell line none 180 311 18
    418 H1299-2 T lung cell line low serum/0.1% FBS low serum mutant 179 0 0
    33 uterus-h N uterus tissue none 179 0 0
    216 BioMarker_BS-2 N endo cell line HUVEC control-1h none 176 822 197
    381 ADR-RES-8 T breast cell line 10 μM cisplatin mutant 174 0 0
    103 NCI-H226 T lung cell line Lung squarnous ca none 173 0 0
    269 SA-OS (Mandy) poly A+ T bone cell line h osteogenic sarcoma, primary none 172 0 24
    63 HT138 T kidney tissue none 172 0 38
    108 U251 T neuro cell line Glioblastoma none 171 1,326 0
    452 EKVX-2 T lung cell line low serum/0.1% FBS low serum mutant 171 0 0
    245 SK-MEL-5 T mel cell line Malignant melanoma, metastasis to none 171 0 62
    axillary node
    154 SNB-75 T neuro cell line Astrocytoma none 170 168 0
    405 AngloTest1-2 T endo cell line HeLa25X DEF-MES for Hypoxia, 1h 25X DEF-MES 169 0 0
    140 SK-MEL-2 T mel cell line Malignant melanoma, metastasis to none 168 751 0
    skin of thigh
    442 Hs68-2 N lung cell line low serum/0.1% FBS low serum wt 166 0 0
    371 SF539-7 T neuro cell line 400 ng/ml noco-24 wt 166 0 0
    275 HT334 T MG tissue none 166 174 0
    80 HT170 T pro tissue none 164 0 0
    263 h fibroblasts Mar. 31, 1992 #12 T fibroblast cell line metastasis to supraorbital area unknown 164 758 117
    416 H1299-1 T lung cell line normal/10% FBS none mutant 163 0 0
    493 Prostate_sampleMG-13 unknown neuro cell line TC-71 Ewings tumor derived cell none 162 0 0
    line
    144 SK-MEL-28 T mel cell line Malignant melanoma none 156 0 0
    126 Caki-1 T renal cell line Clear cell carcinoma, renal primary, none 157 296 0
    metastasis to skin
    326 Hs 578T T breast cell line Dactal carcinoma none 155 0 0
    47 AngloTest1-4 T liver cell line HepG2 25X DEF-MES for Hypoxia, 25X DEF-MES 155 0 0
    0h
    81 HELA-4h-031899 T endo cell line 155 739 0
    492 Prstate_sampleMG-12 unknown neuro cell line TC-32 Ewings tumor derived cell none 152 0 0
    line
    281 pituitary gland-h N neuro tissue none 161 0 218
    404 SW480-3 T col cell line 200 μM minosine mutant 151 0 0
    135 SW-620 T col cell line Colon adenocarcinoma, lymph node none 151 52 318
    metastasis
    181 KB poly A+ T unknown cell line h epidermoid cancer none 148 0 0
    357 HT29-8 T col cell line 400 ng/ml noco-48 mutant 148 0 0
    451 SF-268-3 T neuro cell line 200 μM minosine mutant 147 0 0
    116 CCRF-CEM T LEU cell line ALL Acute lymphobilastic leukemia none 147 1,543 202
    70 HT190 T kidney tissue none 147 676 0
    290 HT312 T lung tissue none 144 0 31
    215 HT371 T lung tissue none 144 75 98
    308 PC-3 T pro cell line Prostate adenocarcinoma none 144 139 0
    372 OVCAR-5-4 T OV cell line 3 mM HU mutant 142 0 0
    32 HMBC N endo cell line coronary artery endothelial cells none 140 986 0
    197 fetal brain-h N neuro tissue none 140 0 0
    222 BioMarker_BS-8 N endo cell line HUVEC 5416-1h SU5416 138 191 0
    296 HT162 T OV tissue none 137 0 0
    2 lymph node-h N heme tissue none 136 3,683 323
    307 PT cells poly A+ T unknown cell line none 133 0 66
    478 AngloTest1-8 N endo cell line HUVEC 10 mm stimulation with VEGF 133 0 0
    VEGF
    258 OVCAR-4-4 T OV cell line 3 mM HU wt 133 0 0
    19 kidney-h N renal tissue none 131 2,148 55
    145 UACC-62 T mel cell line none 130 0 42
    261 neuroblastoma RNA T neuro tissue none 129 0 395
    358 MCF-7-6 T breast cell line 10 μM cisplatin wt 128 0 0
    165 Caki-1 T renal cell line Clear cell carcinoma, renal primary, none 128 134 262
    metastasis is skin
    313 HT192 T MG tissue none 127 656 290
    395 HeLa-6 T endo cell line 10 μM cisplatin HPV E6 127 0 0
    24 stomach-h N col tissue none 124 4,601 47
    295 458 medulla RNA T neuro none 124 0 0
    337 A549-4 T lung cell line 3 mM HU wt 124 0 0
    115 OVCAR-3 T OV cell line Ovary adenocarcinoma none 124 959 0
    225 BioMarker_BS-11 N endo cell line HUVEC VEGF + 5416-1h VEGF 123 0 0
    325 HT395 T lung tissue none 121 0 184
    162 DU-145 T pro cell line Prostate carcinoma none 117 0 16
    125 RXF 393 T renal cell line none 117 212 295
    407 H1299-8 T lung cell line 400 ng/ml noco-48 mutant 116 0 0
    455 SF-268-5 T neuro cell line 2 μM AUR2 inhibitor mutant 116 0 0
    360 OVCAR-4-5 T OV cell line 2 μM AUR2 inhibitor wt 118 0 0
    243 SK-MEL-2 T mel cell line Malignant melanoma, materials to none 113 406 119
    skin of thigh
    199 placenta-h N plac tissue none 113 481 139
    221 HT372-normal N lung tissue none 111 0 59
    334 HCT-118-4 T col cell line 3 mM HU wt 110 0 0
    247 SK-MEL-28 T mel cell line Malignant melanoma none 109 255 0
    125 SR T LEU cell line Large Cell leukemia none 106 0 0
    333 A549-2 T lung cell line low serum/0.1% FBS low serum wt 107 0 0
    477 Prostate_sampleMG-3 unknown pro unknown unknown 106 0 0
    271 MK poly A+ T unknown unknown unknown 105 244 266
    207 CRL1441 + TPA (24h) 8/30 T renal cell line h lung, embryosis TPA 104 796 0
    87 HT348 T pro tissue none 103 1,459 0
    472 AngloTest1-5 T liver cell line HopG2 25X DEF-MES for Hypoxia, 25X DEF-MES 102 0 0
    1h
    385 ADR-RES-8 T breast cell line 400 ng/ml noco-48 mutant 102 0 0
    406 SW480-4 T col cell line 3 mM HU mutant 101 0 0
    59 lung-h N lung tissue none 100 0 123
    160 CCRF-CEM T LEU cell line ALL Acute lymphobllastic leukemia none 100 0 0
    301 lymph node-h N heme tissue none 100 0 144
    152 SNB-19 T neuro cell line Glioblastoma none 100 0 324
    454 EKVX-3 T lung cell line 200 μM minosine mutant 96 0 0
    383 ADR-RES-7 T breast cell line 400 ng/ml noco-24 mutant 97 0 0
    322 TK-10 T renal cell line none 97 0 167
    408 SW480-5 T col cell line 2 μM AUR2 inhibitor mutant 97 0 0
    356 OVCAR-4-3 T OV cell line 200 μM minosine wt 97 0 0
    18 small intestine-h N col tissue none 98 4,224 0
    450 EKVX-1 T lung cell line normal/10% FBS none mutant 96 0 0
    113 HOP-62 T lung cell line Lung adenocarcinoma none 95 0 0
    321 BioMarker_BS-12 N endo cell line HUVEC VEGF + 5416-6h VEGF 95 389 175
    274 testis-h N testes tissue none 94 0 0
    173 Wt-38 72h N lung cell line unknown 94 310 18
    458 EKVX-8 T lung cell line 400 ng/ml noco-48 mutant 94 0 0
    409 C33A-1 T cervical cell line normal/10% FBS none mutant 94 0 0
    198 HCABC N endo cell line none 94 0 0
    95 HT308 T MG tissue none 94 0 0
    302 RPTBC N endo cell line mammary epithelial cells none 94 398 0
    330 HT218-normal N lung tissue none 93 0 57
    467 Prostate_sampleMG-20 unknown pan unknown Hu71 unknown 92 0 0
    124 A498 T renal cell line Kidney carcinoma none 91 0 0
    335 A549-3 T lung cell line 200 μM minosine wt 90 0 0
    38 lymph node-h N heme tissue none 90 636 0
    182 HEPM 34 TGFB1 detergent + DNas N palatal cell line TGFB1 detergent TGFB1 90 0 0
    16 skeletal muscle-h N muscle tissue none 89 3,018 826
    272 Spleen-h N heme tissue none 89 0 205
    132 786-D T renal cell line Primary renal cell adenocarcinoma none 89 0 87
    40 thymus-h N heme tissue none 88 1,145 259
    387 HeLa-1 T endo cell line normal/10% FBS none HPV E6 88 0 0
    349 EKVX-6 T lung cell line 10 μM cisplatin mutant 87 0 0
    56 HT155 T lung tissue none 85 607 108
    395 ADR-RES-2 T breast cell line low serum/0.1% FBS low serum mutant 84 0 0
    473 Prostate_sampleMG-1 unknown pro unknown unknown 82 0 0
    127 DU-145 T pro cell line Prostate carcinoma none 82 0 107
    287 trachea-h N trachea tissue none 80 0 46
    309 brain-h N neuro tissue none 79 58 314
    270 spinal cord-h N neuro tissue none 78 0 71
    238 OVCAR-4 T OV cell line none 76 0 9
    449 SF-368-3 T neuro cell line low serum/0.1% FBS low serum mutant 78 0 0
    189 MCF-7/ADR-RES T breast cell line none 74 0 0
    192 Duodenum-h N col tissue none 74 0 0
    362 OVCAR-4-6 T OV cell line 10 μM cisplatin wt 74 0 0
    71 HT145 T MG tissue none 72 639 0
    239 SN12C T neuro cell line none 71 0 120
    428 U2O6-3 T bone cell line 200 μM minosine mutant 71 0 0
    190 Skeletal muslce-h N muscle tissue none 70 0 0
    182 HOS poly A+ T bone cell line h osteogenic sarcoma none 59 0 0
    298 HT398-normal N lung tissue none 59 0 208
    241 LOX IMVI T mel cell line Arnstanotic melanoma none 57 0 66
    475 Prostate_sampleMG-2 unknown pro unknown unknown 56 0 0
    153 MCF-7/ADR-RES T breast cell line none 55 0 206
    138 Malme-3M T renal cell line Malignant melanoma, metastasis to none 85 104 0
    lung
    90 HELA-8h-031899 T endo cell line 83 1,319 0
    62 HT172 T OV tissue none 83 0 274
    385 MCF-7-5 T breast cell line 2 μM AUR2 inhibitor wt 82 0 0
    47 h adult SMC Oct. 21, 1992 #17 T sme cell line unkinwon 82 585 0
    367 SF639-5 T neuro cell line 2 μM AUR2 inhibitor wt 0 0 0
    374 OVCAR-4-1 T OV cell line normal/10% FBS none wt 0 0 0
    376 OVCAR-4-2 T OV cell line low serum/0.1% FBS low serum wt 0 0 0
    390 MCF-7-7 T breast cell line 400 ng/ml noco-24 wt 0 0 0
    429 Hs68-7 N lung cell line 400 ng/ml noco-24 wt 0 0 0
    431 Hs68-8 N lung cell line 400 ng/ml noco-48 wt 0 0 0
    437 Wt38-3 N lung cell line 200 μM minosine wt 0 0 0
    439 Wr38-4 N lung cell line 3 mM HU wt 0 0 0
    443 Wt38-6 N lung cell line 10 μM cisplatin wt 0 0 0
    444 Hs68-3 N lung cell line 200 μM minosine wt 0 0 0
    445 Wt38-7 N lung cell line 400 ng/ml noco-24 wt 0 0 0
    448 Hs68-4 N lung cell line 3 mM HU wt 0 0 0
    447 Wt38-8 N lung cell line 400 ng/ml noco-48 wt 0 0 0
    460 HCT-116-2 T col cell line low serum/0.1% FBS low serum wt 0 0 0
    488 OVCAR-4-8 T OV cell line 400 ng/ml noco-48 wt 0 0 0
    42 Fetal brain-h N neuro tissue none 0 0 0
    89 HT368 T lung tissue none 0 0 0
    96 HELA-12h-031899 T endo cell line 0 0 0
    100 IRGOV1 T OV cell line none 0 0 0
    131 HCC-2998 T col cell line none 0 0 0
    134 TK-10 T renal cell line none 0 0 0
    147 UACC-257 T mel cell line Malignant melanoma none 0 0 0
    150 NCI-H522 T lung cell line Lung adenocarcinoma none 0 0 0
    177 HT140 T kidney tissue none 0 0 0
    186 testis-h N testes tissue none 0 0 0
    191 UTOS (Mundy) poly a+ T bone cell line none 0 0 0
    211 HT369 T lung tissue none 0 0 0
    212 Ken-4 T lung cell line A549 + 50 ng/ml HGF-24h HGF 0 0 0
    217 HT377 T lung tissue none 0 0 0
    220 BioMarker_BS-7 N endo cell line HUVEC VEGF-24h VEGF 0 0 0
    224 BioMaster_BS-10 N endo cell line HUVEC 5416-24h SU5416 0 0 0
    226 HOP-92 T lung cell line Lung large cell adenocarcinoma none 0 0 0
    230 NCI-H23 T lung cell line Lung adenocarcinoma none 0 0 0
    231 RPMI 8226 T LEU cell line Multiple myeloma none 0 0 0
    235 OVCAR-5 T OV cell line none 0 0 0
    242 IRGOV1 T OV cell line none 0 0 0
    246 SF-529 T neuro cell line Glioblastoma none 0 0 0
    249 UACC-257 T mel cell line Malignant melanoma none 0 0 0
    255 HT279 T MG tissue none 0 0 0
    260 bone marrow-h N heme tissue none 0 0 0
    292 HT149-normal N lung tissue none 0 0 0
    299 Fetal brain-h N neuro tissue none 0 0 0
    311 cerebellum-h N neuro tissue none 0 0 0
    315 HT218 T lung tissue none 0 0 0
    471 Prostate_sampleMG-22 unknown pan unknown Hs399 unknown 0 0 0
    480 AngloTest1-9 N endo cell line HUVEC 30 mm stimulation with VEGF 0 0 0
    VEGF
    481 Prostate_sampleMG-8 unknown pro inknown unknown 0 0 0
    482 AngloTest1-10 N endo cell line HUVEC 30 mm stimulation with PDGF 0 0 0
    PDGF
    484 AngloTest1-11 N endo cell line HUVEC 10 mm stimulation with bFGF 0 0 0
    bFGF
    487 Prostate_sampleMG-9 unknown pro unknwon unknown 0 0 0
    494 Prostate_sampleMG-14 unknown neuro unknown ES7 primary Ewings tumor unknown 0 0 0
    496 Prostate_sampleMG-16 T pro tissue metastasis unknown 0 0 0
    498 Prostate_sampleMG-18 T pro tissue metastasis unknown 0 0 0
    499 Prostate_sampleMG-19 T pro tissue metastasis unknown 0 0 0
  • Table 6, “Multiple Tissue Blot”, contains results of probing a Clontech Multiple Tissue Blot with radioactively labeled probes derived from SGP002 and SGP012. The table lists the tissues on the blot and the values obtained for relative gene expression in each tissue. [0363]
    TABLE 6
    Multiple Tissue Blot
    CIP02
    Tissue ID#NA11_SGP012 ID#NA2_SGPO02
    whole brain 1244 14946
    cerebellum left 3610 22681
    substantia nigra 0 14730
    heart 0 14816
    esophagus 2008 15554
    colon, transverse 1607 20564
    kidney 44 25345
    lung 637 27317
    liver 68 37568
    leukamia, HL-60 0 2204
    fetal brain 0 7572
    cerebral cortex 1178 16874
    cerebellum, right 5201 35351
    accumbens nucleus 0 14985
    aorta 203 13539
    stomach 0 22332
    colon, descending 3812 16311
    skeletal muscle 220 20600
    placenta 497 64169
    pancreas 264 19531
    HeLa S3 0 20564
    fetal heart 649 15777
    frontal lobe 0 11984
    corpus callosum 1972 27350
    thalamus 789 22702
    atrium, left 465 14405
    duodenum 695 20940
    rectum 0 12642
    spleen 0 18882
    bladder 528 22077
    adrenal gland 570 138400
    leukemia, K562 0 7331
    fetal kidney 620 38826
    parletal lobe 492 21242
    amygdala 830 14740
    pituitary gland 1620 41283
    atrium, right 754 8285
    jujenum 2358 21596
    thymus 54 29593
    uterus 1427 18077
    thyroid gland 65 25540
    leukemia, MOLT-4 92 8081
    fetal liver 1189 29080
    occipital lobe 449 17070
    caudate nucleus 334 22638
    spinal cord 556 6385
    ventricle, left 0 9420
    ileum 1002 15704
    peripheral blood leukocyte 1435 15521
    prostate 0 46589
    salivary gland 741 45205
    Burkitt's lymphoma, Raji 0 2497
    fetal spleen 0 24452
    temperal lobe 913 15048
    hippocampus 608 13826
    ventricle, right 811 9938
    ilocecum 0 18970
    lymph node 3497 23227
    testis 10751 33336
    mamary gland 2429 43077
    Burkitt's lymphoma, Daudi 2439 2384
    fetal thymus 797 31519
    paracentral gyrus cerebral 0 16294
    cortex
    medulla oblongata 730 18935
    inter-ventricular septum 0 18269
    appendix 0 23931
    bone marrow 1127 10289
    ovary 437 7103
    colorectal adono-carcinoma, 466 15172
    SW480
    fetal lung 1 26587
    pons 875 12156
    putamen 0 27800
    apex of the heart 311 9897
    colon, ascending 1409 12683
    trachea 1894 22056
    lung carcinoma, A549 1276 15151
  • EXAMPLES
  • The examples below are not limiting and are merely representative of various aspects and features of the present invention. The examples below demonstrate the isolation and characterization of the serine/threonine phosphatases of the invention. [0364]
  • Example 1
  • Identification and Characterization of Protein Phosphatase Genes from Genomic DNA [0365]
  • Materials and Methods [0366]
  • Novel phosphatases were identified from the Celera human genomic sequence databases, and from the public Human Genome Sequencing project (http://www.ncbi.nlm.nih.gov/) using hidden Markov models (HMMRs). The genomic database entries were translated in six open reading frames and searched against the model using a Timelogic Decypher box with a Field programmable array (FPGA) accelerated version of HMMR2.1. The DNA sequences encoding the predicted protein sequences aligning to the MR profile were extracted from the original genomic database. The nucleic acid sequences were then clustered using the Pangea Clustering tool to eliminated repetitive entries. The putative protein phosphatase sequences were then sequentially run through a series of queries and filters to identity novel protein phosphatase sequences. Specifically, the HMMR identified sequences were searched using BLASTN and BLASTX against a nucleotide and amino acid repository containing known human protein phosphatases and all subsequent new protein phosphatase sequences as they are identified. The output was parsed into a spreadsheet to facilitate elimination of known genes by manual inspection. Two models were developed, a “complete” model and a “partial” or Smith Waterman model. The partial model was used to identify sub-catalytic phosphatase domains, whereas the complete model was used to identify complete catalytic domains. The selected hits were then queried using BLASTN against the public nrna and EST databases to confirm they are indeed unique. In some cases the novel genes were judged to be orthologues of previously identified rodent or vertebrate protein phosphatases. [0367]
  • Many of the sequences filed in the provisional patents did not contain the entire coding sequence. Extension of partial DNA sequences to encompass the full-length open-reading frame was carried out by several methods. Iterative blastn searching of the cDNA databases listed in Table 7 was used to find cDNAs that extended the genomic sequences. “LifeGold” databases are from Incyte Genomics, Inc (http://www.incyte.com/). NCBI databases are from the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). All blastn searches were conducted using a blosum62 matrix, a penalty for a nucleotide mismatch of −3 and reward for a nucleotide match of 1. The gapped blast algorithm is described in: Altschul, Stephen F., Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman (1997), “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25:3389-3402). [0368]
  • Extension of partial DNA sequences to encompass the full-length open-reading frame was also carried out by iterative searches of genomic databases. The first method made use of the Smith-Waterman algorithm to carry out protein-protein searches of the closest homologue or orthologue to the partial. The target databases consisted of Genescan and open-reading frame (ORF) predictions of all human genomic sequence derived from the human genome project (HGP) as well as from Celera. The complete set of genomic databases searched is shown in Table 8, below. Genomic sequences encoding potential extensions were further assessed by blastp analysis against the NCBI nonredundant to confirm the novelty of the hit. The extending genomic sequences were incorporated into the cDNA sequence after removal of potential introns using the Seqman program from DNAStar. The default parameters used for Smith-Waterman searches were as shown next. Matrix: blosum 62; gap-opening penalty: 12; gap extension penalty: 2. Genescan predictions were made using the Genescan program as detailed in Chris Burge and Sam Karlin “Prediction of Complete Gene Structures in Human Genomic DNA”, JMB (1997) 268(1):78-94). ORF predictions from genomic DNA were made using a standard 6-frame translation. [0369]
  • Another method for defining DNA extensions from genomic sequence used iterative searches of genomic databases through the Genescan program to predict exon splicing. These predicted genes were then assessed to see if they represented “real” extensions of the partial genes based on homology to related phosphatases. [0370]
  • Another method involved using the Genewise program (http://www.sanger.ac.uk/Software/Wise2/) to predict potential ORFs based on homology to the closest orthologue/homologue. Genewise requires two inputs, the homologous protein, and genomic DNA containing the gene of interest. The genomic DNA was identified by blastn searches of Celera and Human Genome Project databases. The orthologs were identified by blastp searches of the NCBI non-redundant protein database (NRAA). Genewise compares the protein sequence to a genomic DNA sequence, allowing for introns and frameshifting errors. [0371]
    TABLE 7
    Databases used for cDNA-based sequence extensions
    Database Database Date
    LifeGold templates October 2000
    LifeGold compseqs October 2000
    LifeGold compseqs October 2000
    LifeGold compseqs October 2000
    LifeGold fl October 2000
    LifeGold flft October 2000
    NCBI human Ests October 2000
    NCBI murine Ests October 2000
    NCBI nonredundant October 2000
  • [0372]
    TABLE 8
    Databases used for genomic-based sequence extensions
    Database Number of entries Database Date
    Celera v. 1-5 5,306,158 Jan. 19, 2000
    Celera v. 6-10 4,209,980 Mar. 24, 2000
    Celera v. 11-14 7,222,425 Apr. 24, 2000
    Celera v. 15 243,044 May 14, 2000
    Celera v. 16-17 25,885 Apr. 4, 2000
    Celera Assembly 5 (R1.25) 3,313 Oct. 13, 2000
    Celera Assembly 4 (R1.24) 636,234 Aug. 28, 2000
    Celera Assembly 3 (R 1.22, 1,132,900 Jul. 21, 2000
    1.23)
    HGP Phase 0 4,944 May 4, 2000
    HGP Phase 1 28,478 May 5, 2000
    HGP Phase 2 1,508 May 4, 2000
    HGP Phase 3 9,971 May 5, 2000
    HGP Phase 0 3,189 Nov. 1, 2000
    HGP Phase 1 20,447 Nov. 1, 2000
    HGP Phase 2 1,619 Nov. 1, 2000
    HGP Phase 3 9,224 Nov. 1, 2000
    HGP Chromosomal assemblies 2759 Aug. 1, 2000
  • Results: [0373]
  • The sources for the sequence information used to extend the genes in the provisional patents are listed below. For genes that were extended using Genewise, the accession numbers of the protein ortholog and the genomic DNA are given. (Genewise uses the ortholog to assemble the coding sequence of the target gene from the genomic sequence). The amino acid sequences for the orthologs were obtained from the NCBI non-redundant database of proteins .(http://www.ncbi.nlm.nih.gov/Entrez/protein.html). The genomic DNA came from two sources: Celera and NCBI-NRNA, as indicated below. cDNA sources are also listed below. Abbreviations: HGP: Human Genome Project; NCBI, National Center for Biotechnology Information. [0374]
  • SGP006 (SEQ ID NO: 1) [0375]
  • The N-terminal region (1-335) was derived from Genewise predictions using Celera contig 300825903, with protein homologs gi|7242951, gi|8923483 and gi|6714641. Genscan predictions of Celera contig 300825903 was also used. NCBI ESTs used to extend sequence: BE793092.1, gi|9127446, gi|5927364, gi|8148569, gi|9096610, gi|10214290, gi|5927365, gi|4533101, gi|1948748, gi|2010582, gi|30571, gi|2433225, gi|8152915. Incyte sequence 339266.1 is missing exon 7 (GFSVSTAGRMHIFKPVSVQAMW). Public sequence gi|7242951 (KIAA1298) is missing [0376] exon 11 and starts near the beginning of exon 10. The lack of exon 11 causes a frameshift, and so KIAA1298 has a divergent N-terminal predicted peptide, reading exon 10 in a different frame. SGP006 is identical to KIAA1298 over the C-terminal 715 amino acids of SGP006 (amino acids 335 to 1049).
  • [0377]
  • SGP006 (SEQ ID NO: 1) is 6374 nucleotides long. The open reading frame starts at position 34 and ends at position 3183, giving an ORF length of 3150 nucleotides. The predicted protein is 1049 amino acids long. This sequence is fill length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 12q21.3-q22. Amplification of this chromosomal position has been associated with the following human diseases: Bladder carcinoma (12q21-q24, 1/16) (Knuutila, et al.). This gene contains candidate single nucleotide polymorphisms at the following postions: 6222=R (ccaaacataagtggcacar) dbSNP|rs881179_allele. ESTs for this gene in the public domain (dbEST) are: BE793092.1, AI651213.1, BE256978.1. This gene has repetitive sequence at the following nucleotide positions: Alu 5750-6010; 5750-5770. [0378]
  • SGP002 (SEQ ID NO: 2) [0379]
  • SGP002 nucleic acid sequence was derived from Genewise algorithm run with Celera genomic DNA 70000016592596 and the protein homolog gi[0380] 6679156. A similar Genscan prediction gave an N-terminal extension, and comparison with HGP contig gi|7658297 corrected a frameshift in the genewise prediction. Close homologs are of same length. NCBI ESTs gi|7950699 and gi|760983 extend into 5′ and 3′ UTRs respectively. Genomic sequence was used to correct sequence errors in these ESTs. NCBI EST gi|0717958 encodes a splice variant. Incyte EST 1026659.2 encodes an alternative splice form missing an exon which includes part of the phosphatase domain. Incyte EST 1026659.7 adds further 172 nucleotides of 5′ UTR to the gene. Incyte and public ESTs show expression in many tissues, most commonly digestive system, nervous system, respiratory system, and male and female genitalia.
  • SGP002 (SEQ ID NO: 2) is 2732 nucleotides long. The open reading frame starts at position 538 and ends at position 2535, giving an ORF length of 1998 nucleotides. The predicted protein is 665 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 12p11.1-p12.1. This chromosomal position has been associated with the following human diseases: Testis cancer (12p11.2-p12.1, 10/11); non-small cell lung cancer (12p11.2-p12, 4/50), and breast carcinoma (12p11-pter, 2/36) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: BE897795. This gene has repetitive sequence at the following nucleotide positions: 2610-2631. [0381]
  • SGP001 (SEQ ID NO: 3) [0382]
  • Used genscan, and genewise with Celera contig 5000012164505, and protein homologs gi[0383] 6714641 and gi7242951. Several public and Incyte ESTs were used to extend the gene, using genomic data to correct for EST sequence errors. They were: Incyte sequences: 210343.1, 210343.2, 637331CB 1; and NCBI ESTs: gi |3894502, gi|11100172, gi|11100172, gi|4137370, gi|6505071, gi|6885171, gi|1123262, and gi|6590412.
  • SGP001 (SEQ ID NO: 3) is 2260 nucleotides long. The open reading frame starts at position 709 and ends at position 2205, giving an ORF length of 1497 nucleotides. The predicted protein is 498 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position Xp11.1-11.3. This chromosomal position has been associated with the following human diseases: Prostate cancer (Xp11-q13, 1/9) and small cell lung cancer (Xp11.2, 1/13) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: AI272231, BF206586. This gene has repetitive sequence at the following nucleotide positions: 579-598. [0384]
  • SGP018 (SEQ ID NO: 4) [0385]
  • The sequence for SGP018 is predicted from Celera contig 68000017706859, using Gensca and genewise with gi[0386] 7305011 and gi7705959. The genewise prediction covered most of a putative phosphatase. The Genscan prediction overlapped and extended the genewise predictions, and almost all of the genscan was covered by ESTs from Incyte and dbEST. In all cases, ESTs were corrected by first aligning with genomic (Colera/HGP) sequence. A splice variant predicted by Genscan would replace the sequence SEFLDEALLTYR with YCHYIIFSCVFIS (changes the nt sequence ACTGTCATTACATCATTTTCTCTTGTGTTTTCATTTC to CTGAGTTCCTGGATGAGGCGCTGCTGACTTACAG. EST origins: Incyte sequences: 981712.1, 981712.3, 981712.2, 364575.1, 061688.1, 144608.1, 7668648H1, 7473603CB1, 7473604CB1. Public ESTs, including: gi|6880197, gi|6880191, gi|6880141, gi|5441204, gi|5441149, gi|1242174, gi|10984357. Genscan also predicts an alternative C-terminus, where the sequence from VHLL to the C-terminus is replaced by ANGNSVRSTSRFSSSSTREGREMHKFSRSTYNETSSSREESPEPYFFRRTPESSEREESPEPQRP WARSRDWEDVEESSKSDFSEFGAKRKFTQSFMRSEEEGEKERTENREEGRFASGRRSQYRRSNDR EEEEMDDEAIIAAWRRRQEETRTKLQKRRED.
  • The cDNA sequence from 544-612 is not covered by any ESTs. Accordingly, the upstrea and downstream sequences could be different genes and a start at position 613 would give a peptid a later start, at MLESAE; this would give a protein with good homology and the same N-terminal length as the closest mouse homolog, PTP13. A possible alternative splice form seen by comparin incyte ESTs 061688.1 and 7668648H1 predicts a protein form which is missing the Nterminus and instead starts at the sequence MTPEK [0387]
  • SGP018 (SEQ ID NO: 4) is 4361 nucleotides long. The open reading frame starts at position 208 and ends at position 3609, giving an ORF length of 3402 nucleotides. The predicted protein is 1133 amino acids long. This sequence is fill length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene has not been mapped to a chromosomal position. This gene contains candidate single nucleotide polymorphisms at the following postions: 2929=M (agaagatgtctgagtacm) dbSNP|ss1765941; 1161=S (catotaccccaatgas) dbSNP|ss1765940. ESTs for this gene in the public domain (dbEST) are: BF114881. This gene has repetitive sequence at the following nucleotide positions: 1603-1627. [0388]
  • SGP003 (SEQ ID NO: 5) [0389]
  • SGP003 sequence is derived from Genewise with Celera contig 173000019613519 and NCBI homolog template gi[0390] 7705959, extended to the stop codon by genomic walk. The cDNA template is built from 4 EST clones, 2 from muscle, one each from bone and parathyroid gland. Corrected a frameshift in the sequence using HGP contig gi|10178266, and further extended the sequence by 5′ walking the genomic until the first stop. SGP003 has a 235 nucleotide open reading frame preceding the start codon, extending from nucleotide 3 to nucleotide 239, shown in capital letters below:
         caAGGGTTTCAGGTCGCACTGGAAAATCATTTTGCAAGCAGATGT
    CATAGGTCTCCTCTTAGACTGGACGGCACGCAAGGTCAGCGTCACAGATC
    TGACCCTAAAAATAGGCCTCTGTTGCCAGTCGGGGTGGCTGGGCGTGCGG
    CTGCTACATGCCCCACGGACCAGAACCTCCCGACGCGGCCAGGCCCCGGC
    ACACCCAGCTGCAGAAAGGAGAGAAAATCCCTTGGCTCTAAAatg
  • This open reading frame codes for the following peptide sequence: [0391]
  • QGFQVALENHFASRCHRSPLRLDGTQGQRHRSDPKNRPLLPVGVAGRAAATCPTDQ NLPTRPGPGTPSCRKERKSLGSK [0392]
  • The start codon at position 240 conforms to the Kozak rule for initiating methionines, having an A at the −3 position. [0393]
  • SGP003 (SEQ ID NO: 5) is 1262 nucleotides long. The open reading frame starts at position 240 and ends at position 902, giving an ORF length of 663 nucleotides. The predicted protein is 220 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position CHR10. This gene has repetitive sequence at the following nucleotide positions: 311-334. [0394]
  • SGP014 (SEQ ID NO: 6) [0395]
  • Sequence for SGP014 was built from Celera contig 92000005033031 using genscan and genewise, with protein homolog templates gi[0396] 7293532, gi7705959 and gi9502074. The predicted genewise/genscan proteins were extended by overlaps with several ESTs from dbEST (AA723271, AW444890.1, AA435513.1), and confirmed the public sequence gi|7705959. The full predicted peptide is 549 AA, with full DSP domains from 37-181 and 368-541. The following NCBI ESTs come from this gene: gi|11105857, gi|1998334, gi|1423340, gi|2186481, gi|6986652, gi|10372533, gi|2186305, gi|4825880, gi|2740908, gi|3213953, gi|2436350, gi|2140427, gi|2833919, gi|5768154, gi|1134009, gi|2046580, gi|4822411, gi|11152927. The following Incyte Sequences come from this gene: 128077.1, 1384255.1, 8009838H1, 304421CB1. Alternative splicing is very prevalent. The individual exons are as follows: a parenthetical AA at the end of an exon is a residue which crosses the exons at least in the FL form:
    >Exon1:
    MAETSLPELGGEDKATPCPSILELEELLRAGKSSCSRVDEVWPNLFIGD
    (A)
    >Exon2:
    ATANNRFELWKLGITHVLNAAHKGLYCQGGPDFYGSSVSYLGVPAHDLPD
    FDISAYFSSAADFIHRALNTPG (A)
    >Exon3:
    KVLVHCVVGVSRSATLVLAYLMLHQRLSLRQAVITVRQHRWVFPNRGFLH
    QLCRLD (H)
    >Exon4:
    WSLLPAMGLCHFATLALILLVLLEALAQADTQKMVEAQRGVGPRACYSIW
    LLLAPTPPLSHCLQSPQ
    >Exon5:
    KQHQVCGDRRLKASSTNCPSEKCTAWARYSHRW
    >Exon6:
    AHILVPLKIQLRRVPDSFSQQMPETSYLTRVGPDIQCWPESW (G)
    >Exon7:
    MDSLQKQDLRRPKIHGAVQASPYQPPTLASLQRLLWVRQAATLNHIDEVW
    PSLFLGD (A)
    >Exon8:
    YAARDKSKLIQLGITHVVNAAAGKFQVDTGAKFYRGMSLEYYGIEADDNP
    FFDLSVYFLPVARYIRAALSVPQ (E)
    >Exon9:
    DGHGCLFFPKGWVVQGQVADAKLVLPTGRVLVHCAMGVSRSATLVLAFLM
    ICENMTLVEAIQTVQAHRNICPNSGFLRQLQVLDNRLGRETGRF
  • [0397] DSP domain 1 runs from the second half of Exon1 to the end of exon3, domain 2 runs from towards the end of exon7 to almost the end of exon 9. Alternative splicing shown by ESTs: Start of exon 9 (EDG-LPT) is missing in gi|6986652, gi|2186305, gi|10372533 is missing the end of exon 8 and the beginning of exon 9 (KFQ-LPT), gi|11105857 is missing exons 2, 3, 4, 6, and the beginning of exon 9 (EDG-GRV). It has a frameshift between exon 1 and 5, which may be a sequencing error, gi|2186481 is missing exons 2, 3, 6. gi|2740908, gi|2436350, gi|2140427, gi|2833919 have a frameshift relative to the consensus towards the end of exon 9, which replaces the sequence after NSGF with SGSSRFWTTDWGGRRGGSDLAGSQDP*. This change destroys the end of the phosphatase domain, and is not similar to anything in the database. It could be due to genomic polymorphism between individuals, a repeated sequencing error, or possibly some form of gene regulation. These ESTs come from testis (2, same library), prostate and cardiac, so are not a library artifact. 8009838H1 has an internal deletion within exon 2 from YLG-SSA. 304421CB1 is missing exons 2-6 and has a frameshift between exons 1 and 7, and is missing start of exon 9. 128077.1 is missing exons 2,3,6 and the start of exon 9.
  • SGP014 (SEQ ID NO: 6) is 1917 nucleotides long. The open reading frame starts at position 31 and ends at position 1680, giving an ORF length of 1650 nucleotides. The predicted protein is 549 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 10q21.3. This chromosomal position has been associated with the following human diseases: Squamous cell carcinomas of the head and neck (10q21-q22, 2/30) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: AA723271, AW444890.1, AA435513.1. [0398]
  • SGP060 (SEQ ID NO: 7) [0399]
  • The sequence of SGP060 is derived from Genewise, using Celera contig 6514035[0400] —1 and protein homolog NP057448. NCBI ESTs used to extend the sequence include BF207232, BF314818, AW953216.1.
  • SGP060 (SEQ ID NO: 7) is 636 nucleotides long. The open reading frame starts at [0401] position 1 and ends at position 636, giving an ORF length of 636 nucleotides. The predicted protein is 211 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, MKP. This gene maps to chromosomal position 8p11.1-q11.1 centromeric. This chromosomal position has been associated with the following human diseases: breast carcinoma (8p11-p12, 8/53); non-small cell lung cancer (18p11.2, 2/50) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: BF207232, BF314818, AW953216.1.
  • SGP008 (SEQ ID NO: 8) [0402]
  • Genscan and genewise were done on Celera contig 78000006091415, using homologs gi|9910432, gi|7294466 and gi|7298988. These were verified and extended with public ESTs gi|7280554, gi|6925677 and gi|6142140, and Incyte sequence 7475576CB1. The predicted cDNA was corrected using sequence from the Celera contig and current HGP contigs. Comparison with non-human ESTs and public protein sequences indicate that there may be an internal start to the protein, at amino acid position 95 (at MGNG). [0403]
  • SGP008 (SEQ ID NO: 8) is 1326 nucleotides long. The open reading frame starts at [0404] position 1 and ends at position 990, giving an ORF length of 990 nucleotides. The predicted protein is 329 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Dual Phosphatase, DSP, STYX. This gene maps to chromosomal position 20q11.2. This gene contains candidate single nucleotide polymorphisms at the following postions: 871=S (cagcagcctccgagggaaccs) dbSNP|ss1389419. ESTs for this gene in the public domain (dbEST) are: AW406620.1, BF377364.1, AW593296.1. This gene has repetitive sequence at the following nucleotide positions: 1251-1270.
  • SGP039 (SEQ ID NO: 9) [0405]
  • SGP039 is derived from Celera sequence 17000030279756, and from Incyte sequences 272616.1 and 7476908CB1. [0406]
  • SGP039 (SEQ ID NO: 9) is 1083 nucleotides long. The open reading frame starts at [0407] position 1 and ends at position 1083, giving an ORF length of 1083 nucleotides. The predicted protein is 360 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Serine Phosphatase, STP, PP2C. This gene has not been mapped to a chromosomal position. ESTs for this gene in the public domain (dbEST) are: BE147139.
  • SGP040 (SEQ ID NO: 10) [0408]
  • The sequence for SGP040 is derived from Celera sequence 17000091609039 and the public sequence NM[0409] 018444.1 for pyruvate dehydrogenase phosphatase.
  • SGP040, PDP (SEQ ID NO: 10) is 1725 nucleotides long. The open reading frame starts at [0410] position 1 and ends at position 1725, giving an ORF length of 1725 nucleotides. The predicted protein is 574 amino acids long. This sequence is full length (start methionine to stop codon). It is classified as (superfamily/group/family): Serine Phosphatase, STP, PP2C. This gene maps to chromosomal position 8q21.3. This chromosomal position has been associated with the following human diseases: Mantle cell lymphoma (18q21-q23; 5/50) (Knuutila, et al.). ESTs for this gene in the public domain (dbEST) are: AV706533.1, AV705571.1, AV710801.1.
  • SGP012 (SEQ ID NO: 11) [0411]
  • The sequence for SGP012 is derived from Genewise, using Celera sequences 94000002120453; 142000016367225; 142000016006753, as geneomic DNA input and NP[0412] 031981 (murine PTP-EST) as protein homolog. Incyte ESTSs that overlap this sequence include 1005303.1, and 71096513. Public ESTs which overlap with the sequence include AL042532.1, AI381571, and AW872677.
  • SGP012 PTP-ESP (SEQ ID NO: 11) is 4719 nucleotides long. The open reading frame starts at [0413] position 1 and ends at position 4719, giving an ORF length of 4719 nucleotides. The genomic sequence for this gene is of fairly poor quality, i.e., it has not been assembled and has apparent sequence errors. Thus the nucleic acid and protein sequences are partial, with gaps indicated by “X” s in the sequence. The predicted protein is 1573 amino acids long. This sequence contains the catalytic domain. It is classified as (superfamily/group/family): Tyrosine Phosphatase, RPTm, PTPd. This gene has not been mapped to chromosomal position. ESTs for this gene in the public domain (dbEST) are: AL042532.1, AI381571, AW872677. This gene has repetitive sequence at the following nucleotide positions: 1305-1324.
  • SGP024 (SEQ ID NO: 12) [0414]
  • SGP024 is derived from Genewise using Celera DNA sequence 142000016226692 as geneomic source and NP[0415] 002830.1 (human PTP delta) as protein homolog.
  • SGP024 (SEQ ID NO: 12) is 354 nucleotides long. The open reading frame starts at [0416] position 1 and ends at position 357, giving an ORF length of 357 nucleotides. The predicted protein is 118 amino acids long. This sequence is a partail catalytic domain. It is classified as (superfamily/group/family): Tyrosine Phosphatase, Receptor PTP, PTPdelta sub-family.
  • Example 2
  • Predicted Proteins [0417]
  • SGP006, KIAA1298 (SEQ ID NO: 1) encodes SEQ ID NO: 13, a protein that is 1049 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov profile for a MKP/DSP phosphatase from [0418] profile position 1 to profile position 173 (full length catalytic domain). The position of the catalytic region within the encoded protein is from amino acid 308 to amino acid 446. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results. The C-terminus of SGP006 (amino acid positions 322 to 1049) is 100% identical to KIAA1298 protein [Homo sapiens]. The output can be summarized as follows: P-value=0; number of identical amino acids=715; percent identity=100%; percent similarity=100%; the accession number of the most similar entry in NRAA is BAA92536.1; the name or description, and species, of the most similar protein in NRAA is: KIAA1298 protein [Homo sapiens]. The region N-terminal to this identity with KIAA1298 is novel—for amino acids 120 to 477, the results of a Smith Waterman search of the public database of amino acid sequences (NRAA) yielded the following results: P-value=1.50E-99; number of identical amino acids=248; percent identity=46%; percent similarity=59%; the accession number of the most similar entry in NRAA is BAA89534.1; the name or description, and species, of the most similar protein in NRAA is: MAP kinase phosphatase [Drosophila melanogaster]. The N-terminal sequence of SGP006, from amino acid 1 to 263, is also novel, and the results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=6.80E-58; number of identical amino acids=119; percent identity=41%; percent similarity=59%; the accession number of the most similar entry in NRAA is NP060327.1; the name or description, and species, of the most similar protein in NRAA is: Hypothetical protein FLJ20515 [Homo sapiens].
  • SGP002 (SEQ ID NO: 2) encodes SEQ ID NO: 14, a protein that is 665 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov MKP/DSP phosphatase domain from [0419] profile position 1 to profile position 173 (full length catalytic domain). The position of the catalytic region within the encoded protein is from amino acid 158 to amino acid 297. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=1.10E-157; number of identical amino acids=304; percent identity=46%; percent similarity=60%; the accession number of the most similar entry in NRAA is NP004411.1; the name or description, and species, of the most similar protein in NRAA is: dual specificity phosphatase 8 [Homo sapiens]. This protein contains a Rhodanese-like domain (amino acids 11 to 131). The rhodanese domain has been associated with thiosulfate: cyanide sulfurtransferase (EC 2.8.1.1) activity. The presence of this domain may indicate that SGP002 is regulated in response to the cellular redox environment (Nandi et al., Int J Biochem Cell Biol April 2000; 32(4):465-73; Rhodanese as a thioredoxin oxidase).
  • SGP001 (SEQ ID NO: 3) encodes SEQ ID NO: 15, a protein that is 498 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov profile for a MKP/DSP phosphatase domain from [0420] profile position 1 to profile position 173. The position of the catalytic region within the encoded protein is from amino acid 307 to amino acid 441. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: Pscore=8.30E-133; number of identical amino acids=250; percent identity=47%; percent similarity=60%; the accession number of the most similar entry in NRAA is BAA89534.1; the name or description, and species, of the most similar protein in NRAA is: MKP [Drosophila melanogaster].
  • SGP018 (SEQ ID NO: 4) encodes SEQ ID NO: 16, a protein that is 1133 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov profile from profile position MKP/DSPphosphatase domain from [0421] profile position 1 to profile position 173. The position of the catalytic region within the encoded protein is from amino acid 185 to amino acid 330. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=2.20E-27; number of identical amino acids=79; percent identity=45%; percent similarity=63%; the accession number of the most similar entry in NRAA is NP057448.1; the name or description, and species, of the most similar protein in NRAA is: Protein phosphatase LOC51207 [Homo sapiens].
  • SGP003 (SEQ ID NO: 5) encodes SEQ ID NO: 17, a protein that is 220 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov profile from profile position MKP/DSPphosphatase domain from [0422] profile position 1 to profile position 173. The position of the catalytic region within the encoded protein is from amino acid 54 to amino acid 199. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=3.40E-54; number of identical amino acids=91; percent identity=49%; percent similarity=68%; the accession number of the most similar entry in NRAA is NP057448.1; the name or description, and species, of the most similar protein in NRAA is: Protein phosphatase LOC51207 [Homo sapiens].
  • SGP014 (SEQ ID NO: 6) encodes SEQ ID NO: 18, a protein that is 549 amino acids long, with two phosphatase domains. Both domains in this protein match the hidden Markov profile for an MKP/DSP phosphatase profile from [0423] position 1 to profile position 173 (fall length). Both DSP domains are similar, with best hits to gi|7705959 (human; partial of this gene), and DSP13 from mouse. The position of the catalytic regions within the encoded protein are from amino acid 37 to amino acid 181 for the N-terminal domain, and from 368 to 520 for the C-terminal domain. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: for amino acid 324-549, P-value=7.50E-122; number of identical amino acids=198; percent identity=88%; percent similarity=88%; the accession number of the most similar entry in NRAA is NP057448.1; the name or description, and species, of the most similar protein in NRAA is: Protein phosphatase LOC51207 [Homo sapiens]. For amino acids 1-198, the results of a Smith Waterman search of the public database of amino acid sequences yielded the following results: P-value=8.20E-36; number of identical amino acids=75; percent identity=45%; percent similarity=65%; the accession number of the most similar entry in NRAA is NP004081.1; the name or description, and species, of the most similar protein in NRAA is: Dual specificity phosphatase 3 [Homo sapiens].
  • SGP060 (SEQ ID NO: 7) encodes SEQ ID NO: 19, a protein that is 211 amino acids long. It is classified as an MKP. The phosphatase domain in this protein matches the hidden Markov profile for MKP/DSP phosphatase from [0424] profile position 1 to profile position 173 (full length). The position of the catalytic region within the encoded protein is from amino acid 61 to amino acid 204. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=1.10E-48; number of identical amino acids=86; percent identity=53%; percent similarity 72%; the accession number of the most similar entry in NRAA is NP057448.1; the name or description, and species, of the most similar protein in NRAA is: Protein phosphatase LOC51207 [Homo sapiens].
  • SGP008 (SEQ ID NO: 8) encodes SEQ ID NO: 20, a protein that is 329 amino acids long. It is classified as an MKP./STYX,. The phosphatase domain in this protein matches the hidden Markov profile from profile position MKP/DSPphosphatase domain from [0425] profile position 1 to profile position 173. The position of the catalytic region within the encoded protein is from amino acid 98 to amino acid 235. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=4.40E-172; number of identical amino acids=260; percent identity=92%; percent similarity=92%; the accession number of the most similar entry in NRAA is CAC10008.1; the name or description, and species, of the most similar protein in NRAA is: Novel protein [Homo sapiens].
  • SGP039 (SEQ ID NO: 9) encodes SEQ ID NO: 21, a protein that is 360 amino acids long. It is classified as: PP2C,. The phosphatase domain in this protein matches the hidden Markov profile from [0426] profile position 1 to profile position 301 (Full length catalytic). The position of the catalytic region within the encoded protein is from amino acid 91 to amino acid 344. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=1.00E-106; number of identical amino acids=164; percent identity=98%; percent similarity=99%; the accession number of the most similar entry in NRAA is AAD17235.1; the name or description, and species, of the most similar protein in NRAA is: PP 2C [Mus musculus].
  • SGP040, PDP (SEQ ID NO: 10) encodes SEQ ID NO: 22, a protein that is 574 amino acids long. It is classified as: PP2C. The phosphatase domain in this protein matches the hidden Markov profile from [0427] position 1 to position 301. The position of the catalytic region within the encoded protein is from amino acid 209 to amino acid 497. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=0; number of identical amino acids=574; percent identity=100%; percent similarity=100%; the accession number of the most similar entry in NRAA is NP060914.1; the name or description, and species, of the most similar protein in NRAA is: Pyruvate dehydrogenase phosphatase [Homo sapiens].
  • SGP012 PTP-ESP (SEQ ID NO: 11) encodes SEQ ID NO: 23, a protein that is 1573 amino acids long. It is classified as: PTP, delta phosphatase-like. The phosphatase domain in this protein matches the hidden Markov profile for a PTP phosphatase, from [0428] profile position 1 to profile position 264 (full length catalytic). The position of the catalytic region within the encoded protein is from amino acid 1010 to 1259. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=0; number of identical amino acids=1053; percent identity=60%; percent similarity=70%; the accession number of the most similar entry in NRAA is NP031981.1; the name or description, and species, of the most similar protein in NRAA is: Embryonic stem cell phosphatase [Mus musculus]. This protein contains five fibronectin domains at amino acid positions 35-120; 128-208; 390-471; 484-558; 668-748. Gaps with the sequence are indicted by “XXX”.
  • SGP024 (SEQ ID NO: 12) encodes SEQ ID NO: 24, a protein that is 118 amino acids long. It is classified as a PTP, related to PTP delta. The phosphatase domain in this protein matches the hidden Markov profile for a PTP from profile position 205 to profile position 264 (this is a partial catalytic domain, representing the C-terminal region). The position of the catalytic region within the encoded protein is from [0429] amino acid 3 to amino acid 58. The results of a Smith Waterman search of the public database of amino acid sequences (NRAA) with this protein sequence yielded the following results: P-value=5.90E-54; number of identical amino acids=90; percent identity=76%; percent similarity=82%; the accession number of the most similar entry in NRAA is CAA38068.1; the name or description, and species, of the most similar protein in NRAA is: Protein-tyrosine phosphatase delta [Homo sapiens].
  • Example 3
  • Expression Analysis of Novel Mammalian Protein Phosphatases [0430]
  • The gene expression patterns for selected genes were studied using two techniques: 1) a tissue microarray developed at Sugen, containing 499 tissues and probed with labeled genes; and 2) a commercial array of tissue from Clontech, probed with labeled genes. [0431]
  • 1) Tissue Arrays [0432]
  • “cDNA libraries” derived from a variety of sources were immobilized onto nylon membranes and probed with 32P-labeled cDNA fragments derived from the gene(s) of interest. The sources of RNA are listed in Table 3. They are: 1) Biochain Institute (Hayward, Calif.; http://www.biochain.com/main[0433] 3.html): 2) Clontech (Palo Alto, Calif., http://www.clontech.com/); 3) mammalian cell lines used by the National Cancer Institute (NCI) Developmental Therapeutics Program (http://dtp.nci.nih.gov/; can be ordered from ATCC: http://www.atcc.org/catalogs.html): 4) PathAssociates http://www.saic.com/company/subsidiaries/pai.html; San Diego, Calif.). The protocols for preparing cDNA arrays are detailed below. Several cell lines were treated with compounds to evaluate their effects on gene expression. There were eight treatments: 1) control, 2) low sereum, 3) 200 uM mimosine, 4) 3 mM HU, 5) 2 uM AUR2 inhibitor,) 10 uM cisplatin, 7) 400 ng/ml nocodozole-24 hours, and 8) 400 ng/ml nocodozole-48 hours. The treated cell lines are listed by cell line name followed by a number from 1 to 8.
  • “cDNA libraries” derived from over 450 tissue or cell line sources were immobilized onto nylon membranes and probed with 32P-labeled cDNA fragments derived from the gene(s) of interest. To make the cDNA, total RNA or mRNA was used as template in a reverse transcription reaction to generate single-stranded cDNAs (ss cDNA) that were tagged with specific sequences at each end. An oligo dT primer containing a specific sequence (CDS: AAGCAGTGGTAACAACGCAGAGTACT[0434] 30VN (V=A,G,C N=A,G,C,T)) anneals at the polyA track at the 3′ end of the “mRNA and the reverse transcriptase (MMLV RnaseH) transcribes the antisense strand until it reaches the end of the RNA strand when it adds additional C residues. If a primer (SMII: AAGCAGTGGTAACAACGCAGAGTACGCGGG or ML2G: AAGTGGCAACAGAGATAACGCGTACGCGGG) ending with 3 Gs is added, it anneals to the added Cs and the MMLV recognizes the rest of the primer sequence as template and continues transcription. As a result, the synthesized cDNAs contain specific sequence tags at both the 5′ and the 3′ end. When the 5′ and the 3′ ends are tagged with the same sequence (CDS and SMII) it is referred to as “symmetric”. When the 5′ end is tagged with a different sequence than the 3′ end (CDS and ML2G) is referred to as “asymmetric”. A double-stranded “cDNA library” is then generated by PCR amplification using the 3′PCR and ML2 primers (3′ PCR: AAGCAGTGGTAACAACGCAGAGT and ML2: AAGTGGCAACAGAGATAACGCGT) that anneal to the added sequence tags.
  • The amplified “cDNA libraries” were manually arrayed onto nylon membranes with a 384 pin replicator. The DNA was denatured by alkali treatment, neutralized and cross-linked by UV light. The arrays were pre-hybridized with Express Hyb (Clontech) and hybridized with [0435] 32p labeled probes generated by random hexamer priming of cDNA fragments corresponding to the genes of interest. After washing, the blots were exposed to phosphorimaging cassettes and the intensity of the signal was quantified. The amount of the DNA on the arrays was also quantified by treating non-denatured or denatured arrays with Syber Green I or Syber Green II respectively (1:100,000 in 50 mM Tris, pH8.0) for 2 minutes. After washing with 50 mM Tris, pH8.0, the fluorescent emission was detected with a phosphorimager (Molecular Dynamics) and quantified. The amount of the arrayed DNA was used to normalize the hybridization signal and the corrected values are tabulated in Table 5.
  • Statistical Methods: [0436]
  • The tissue array data for the 3 phosphatases were standardized for statistical analysis across the different tissue types using range standardization. Standardization converts measurements to a common scale. We used range standardization, which subtracts the smallest value of each variable from each value and divides by its range. The new scale starts at 0 and ends at 1.0. The following statistical procedures were implemented on the standardized data: generation of descriptive statistics, graphical visualization, hierarchical and k-means cluster analysis (at 10, 7, and 5 clusters), and comparison of groups using analysis of variance (ANOVA). When tissue-specific data were present for both normal and tumor samples, the two groups were directly compared for fold differences. All statistical analyses were carried out separately for the symmetric and asymmetric tissue array laboratory methods because we know from experience with past data that gene expression is dependent upon the method used. All statistical analyses were carried out using SYSTAT 9.01 (Copyright © 1999 by SPSS, Inc.). [0437]
  • SUMMARY OF RESULTS
  • [0438]
    TABLE 9
    Fold difference in mean expression between normal human tissue
    and cancer cell lines, and between and normal tissue and tumor samples.
    Normal vs. Tumor
    Tissue vs. Cell Within Cell
    CDNA Gene Line Pooled Line Within Tissue
    Symmetric SGP003 23.02 28 6.26 10.92
    Symmetric SGP060 2.86 2.67 −1.92 0
    Symmetric SGP018 2.25 2.33 2.4 −1.32
    Asymmetric SGP003 +2.33** 1.37 1.19 −1.03
    Asymmetric SGP060 +2.38**** 1.08 −2.06 −1.26
    Asymmetric SGP018 1.01 1.51*** −1.43* −1.72**
  • Discussion [0439]
  • 1. SGP003 (SEQ ID NO: 5) [0440]
  • This gene was observed to express consistently higher in tissue samples (as versus cell-line samples) and in normal samples (as versus tumor samples) in both the symmetric and asymmetric methods. We observed much higher fold differences in the symmetric method than in the asymmetric method (Table 9), but because of inadequate sample size and large variation in the data, we did not find the difference to be statistically significant in the symmetric method. On the other hand, the fold difference of 2.33 between the tissue and cell-line samples in the asymmetric method was statistically significant at p<0.05. Because this phosphatase is expressed higher in normal than in tumor samples, it may play a role in tumor suppression. Highest levels of expression of this gene were observed in the normal samples, particularly those drawn from brain, fetal brain, fetal kidney, and glandular tissues such as the pituitary and adrenal gland. We did observe some relatively high levels of expression in a few tumor samples (lymphoblastoma, neuroblastoma, melanoma, lung, colon, breast, and renal tumors). Selected clusters and their rankings according to levels of expression for this phosphatase are listed below: [0441]
  • (Symmetric Data) [0442]
  • Cluster ranking by highest mean expression: [0443]
  • Cluster 1 (singleton). NORMAL GROUP: heart sample (tissue). [0444]
  • Cluster 2 (singleton). NORMAL GROUP: spinal cord (tissue). [0445]
  • Cluster 3 (8 members). NORMAL GROUP only: colon (stomach tissue), colon (small intestinal tissue), mammary epithelial cells (cell line), spleen (heme tissue), lymph node theme tissue), fetal lung (tissue), fetal brain (neural tissue), and prostate (tissue). [0446]
  • (Asymmetric Data) [0447]
  • Cluster ranking by highest mean expression: [0448]
  • Cluster 1 (singleton). NORMAL GROUP: adrenal gland (tissue). [0449]
  • Cluster 2 (3 members). NORMAL GROUP: thymus (heme tissue). TUMOR GROUP: lung carcinoma (cell line), and a neuro sample (tissue). [0450]
  • Cluster 3 (14 members). NORMAL GROUP: thyroid gland (tissue), lymph node (heme tissue), and coronary artery endothelial cells (cell line); and TUMOR GROUP: lung (tissue), malignant melanoma metastasis to lung (cell line), breast (cell line), unknown (cell line), breast (cell line), HNS (tissue), endothelial (cell line), endothelial (cell line), prostate (tissue), kidney (tissue), and renal adenocarcinoma (cell line). [0451]
  • 2. SGP060 (SEQ ID NO: 7) [0452]
  • The highest expressers in the asymmetric method were tumor samples. Although they represented different types of tumors, we observed consistently very high expression in various lung cancer samples. This gene may be an oncogene important in lung cancer. In normal tissues, it expressed highest in brain tissue samples and in fetal kidney. Selected clusters and their rankings according to levels of expression for this phosphatase are listed below: [0453]
  • Cluster ranking by highest mean expression (Asymmetric data): [0454]
  • Cluster 1 (2 members). TUMOR GROUP only: lung (tissue) and lung carcinoma (cell line). [0455]
  • Cluster 2 (3 members). TUMOR GROUP: lung (tissue) and ovary adenocarcinoma (cell line); and NORMAL GROUP: prostate (tissue). [0456]
  • Cluster 3 (3 members). TUMOR GROUP only: lung (tissue), neuroblastoma (tissue), and colon carcinoma (cell line). [0457]
  • Cluster 4 (8 members). TUMOR GROUP: MG (tissue), smc (cell line), glioblastoma (cell line), lung large cell carcinoma (cell line), END (tissue), primary renal cell carcinoma (cell line), and lung (tissue); and NORMAL GROUP: brain (tissue). [0458]
  • Cluster 5 (10 members). TUMOR GROUP: lung (tissue), malignant melanoma metastasis to lung (cell line), colon adenocarcinoma (cell line), renal (cell line), unknown sample (MK ploy A+), breast (cell line), renal primary clear cell carcinoma metastasizing (cell line), ovary (tissue), and neuroblastoma (keratinocyte cell line); and NORMAL GROUP: fetal kidney (tissue). [0459]
  • 3. SGP018 (SEQ ID NO: 4) [0460]
  • According to the asymmetric method, this gene expresses higher in tumor samples (as versus the normal samples) and this pattern was consistent and statistically significant for pooled, within tissue, and within cell-line samples (Table 9). This gene expresses very highly across a broad range of tumor types, and may be particularly important in glioblastoma and ovarian cancer. Like KAP, this phosphatase may be a good target as a marker and in therapeutics. [0461]
  • Cluster ranking by highest mean expression (Asymmetric data): [0462]
  • Cluster 1 (singleton). TUMOR GROUP: neuro (tissue). [0463]
  • Cluster 2 (3 members). TUMOR GROUP only: HNS (tissue), renal adenocarcinoma (cell line), and ovary (cell line). [0464]
  • Cluster 3 (5 members). TUMOR GROUP only: malignant melanoma, metastasis to lung (cell line), colon (cell line, treated with 3 mM HEW), ovary (cell line, treated with 10 uM cisplatin), neuro (cell line, treated with 10 uM cisplatin), and PML peripheral blood, promyelocytic leukemia (cell line). [0465]
  • Cluster 4 (11 members). TUMOR GROUP: colon (cell line, treated with 10 uM cisplatin), breast (cell line), endothelial cells (cell line, treated with HeLa25X DEF-MES for hypoxia, 4 hours), unknown sample (unknown), cervical (cell line, treated with 400 ng/ml noco-48 hours), kidney (tissue), lung (tissue), lung (tissue), endothelial cells (cell line), and lung (tissue); and NORMAL GROUP: HUVEC (cell line, treated with 10 mn PDGF stimulation). [0466]
  • Cluster 5 (27 members). TUMOR GROUP: kidney carcinoma (cell line), lung (tissue), neuro (cell line, treated with 10 uM cisplatin), lung (tissue), bone (cell line), breast (cell line), lung (tissue), lung (cell line, treated with 3 mM HU), neuro (cell line, treated with 400 ng/ml noco-24 hours), endothelial cells (cell line, treated with HeLa25X DEF-MES for hypoxia, 0 hours), ovary (cell line, treated with 2 uM AUR2 inhibitor), breast (cell line, treated with normal/10% FBS), breast (cell line, treated with 2 uM AUR2 inhibitor), breast (cell line, treated with 200 uM mimosine), bone (cell line, treated with low serim/0.1% EBS), colon (cell line, treated with 10 uM cisplatin), cervical (cell line, treated with low serim/0.1% FBS), endothelial cells (cell line), kidney (tissue), pancreas (tissue), and renal (cell lie); NORMAL GROUP: endothelial cells (cell line, treated with HUVEC VEGF+5416-24 hours), lung (tissue), endothelial cells (cell line, HUVEC unstimulated/control), and stomach (colon tissue). [0467]
  • 2) Multiple Tissue Expression Blots (MTE) [0468]
  • MTE (Multiple Tissue Expression) blots were obtained from Clontech Laboratories, Inc (see table 6). These blots contained 84 arrayed cDNA samples derived from normal human tissue and human cell lines, and controls. The expression blots were prehybridized with ExpressHyb hybridization solution (Clontech Laboratories) containing 0.1 mg/ml denatured salmon sperm DNA at a temperature of 65° C. for two hours. Radioactive DNA probes were prepared using the Random Priming DNA labeling kit (Roche). Purified DNA fragments (100 ng) were labeled with 250 uCi of 32P-labeled dCTP for 45 minutes using the kit protocol. Unincorporated nucleotide was removed through the use of a spin column (ProbeQuant G50 micro columns, Amersham Pharmacia, Inc.). After denaturation by boiling for three minutes, the probe was introduced into the prehybridization solution, and the blot was hybridized at 65° C. for 20 hours. The blot was subsequently washed four times for 15 minutes each at 65° C. in a solution containing 15 mM NaCl, 1.5 mM Na[0469] 3Citrate, 0.1% sodiumn lauryl sulfate (SDS) and exposed to the phosphoimager screen for quantitation.
  • Results [0470]
  • SGP012 (SEQ ID NO: 11, encoding SEQ ID NO: 23) is expressed at the highest levels in the following tissues: testis; cerebellum, right; colon, descending; cerebellum left; lymph node; Burkitt's lymphoma; Daudi; and mammary gland. This pattern of expression suggests that SGP012 may play a role in diseases of the central nervous system (cerebellum exprssion), in immune system disease (the lymph node, Burkitt's lymphoma, and Daudi are all immune system tissues), or breast cancer (from expression in mammary tissue). [0471]
  • SGP002 (SEQ ID NO: 2, encoding SEQ ID NO: 14) is expressed at the highest levels in the following tissues: adrenal gland; placenta; prostate; salivary gland; mamary gland; pituitary gland. Expression in the prostate and breast may indicate a role for this phosphatase in cancer of these tissues. Expression in the adrenal gland may indicate a role in metabolic processes controled by that gland, such as stress response. [0472]
  • Example 4
  • Chromosomal Localization of Mammalian Protein Phosphatases [0473]
  • Several sources were used to find information about the chromosomal localization of the genes in the present invention. First, the accession number for the nucleic acid sequence was used to query the Unigene database. The site containing the Unigene search engine is: http://www.ncbi.nlm.nih.gov/UniGene/Hs.Home.html. Information on map position within the Unigene database is imported from several sources, including the Online Mendelian Inheritance in Man (OMIM, http://www.ncbi.nlm.nih.gov/Omim/searchomim.html), The Genome Database (http://gdb.infobiogen.fr/gdb/simpleSearch.html), and the Whitehead Institute human physical map (http://carbon.wi.mit.edu:8000/cgi-bin/contig/sts_info?database=release). If Unigene has not mapped the EST, then the nucleic acid for the gene of interest is used as a query against databases, such as dbsts and htgs (described at http://www.ncbi.nlm.nih.gov/BLAST/blast_databases.html) containing sequences that have been mapped already. The nucleic acid sequence is searched using BLAST-2 at NCBI (http://www.ncbi.nlm.nih.gov/cgi-bin/BLAST/nph-newblast) and is used to query either dbsts or htgs. Once a cytogenetic region has been identified by one of these approaches, disease association is established by searching OMIM with the cytogenetic location. OMIM maintains a searchable catalog of cytogenetic map locations organized by disease. A thorough search of available literature for the cytogenetic region is also made using Medline (http://www.ncbi.nlm.nih.gov/PubMed/medline.html). References for association of the mapped sites with chromosomal abnormalities found in human cancer can be found in: Knuutila, et al., Am J Pathol, 1998, 152:1107-1123. The results are discussed in the Section on Nucleic Acids above. [0474]
  • Example 5
  • Candidate Single Nucleotide Polymorphisms (SNPs) [0475]
  • Materials and Methods [0476]
  • The most common variations in human DNA are single nucleotide polymorphisms (SNPs), which occur approximately once every 100 to 300 bases. Because SNPs are expected to facilitate large-scale association genetics studies, there has recently been great interest in SNP discovery and detection. Candidate SNPs for the genes in this patent were identified by blastn searching the nucleic acid sequences against the public database of sequences containing documented SNPs (dbSNP, at NCBI, http://www.ncbi.nlm.nih.gov/SNP/snpblastpretty.html). dbSNP accession numbers for the SNP-containing sequences are given. SNPs were also identified by comparing several databases of expressed genes (dbEST, NRNA) and genomic sequence (i.e., NRNA) for single basepair mismatches. The results are shown in Table 2, in the column labeled “SNPs”. These are candidate SNPs—their actual frequency in the human population was not determined. The code below is standard for representing DNA sequence: [0477]
    G = Guanosine
    A = Adenosine
    T = Thymidine
    C = Cytidine
    R = G or A, puRine
    Y = C or T, pYrimidine
    K = G or T, Keto
    W = A or T, Weak (2 H-bonds)
    S = C or G, Strong (3 H-bonds)
    M = A or C, aMino
    B = C, G or T (i.e., not A)
    D = A, G or T (i.e., not C)
    H = A, C or T (i.e., not G)
    V = A, C or G (i.e., not T)
    N = A, C, G or T, aNy
    X = A, C, G or T
  • [0478]
    complementary G A T C R Y W S K M B V D H N X
    DNA       +−+−+−+−+−+−+−+−+−+−+−+−+−+−+−+
    strands C T A G Y R S W M K V B H D N X
  • For example, if two versions of a gene exist, one with a “C” at a given position, and a second one with a “T: at the same position, then that position is represented as a Y, which means C or T. In table 1, for SGP002, the SNP column says “1165=R”, which means that at position 1165, a polymorphism exists, with that position sometimes containing a G and sometimes an A (R represents A or G). SNPs may be important in identifying heritable traits associated with a gene. [0479]
  • Results [0480]
  • SGP006 has a single nucleotide polymorphism at position 6222: 6222=R (ccaaacataagtggcacar). The dbSNP accession number is rs881179. This SNP occurs in the 3′ untranslated region. [0481]
  • SGP018 has a single nucleotide polymorphism at position 1161; 1161=S (catctaccccaatgas). The dbSNP accession number is ss1765940. This SNP results in a change in the peptide sequence: amino acid number 183 can be either a glutamic acid, when nucleotide 549=G; or amino acid 183 can be an aspartic acid, when nucleotide 549=C. This change is fairly conservative, since both amino acids are acidic, but could alter the biology of the enzyme. A second SNP is silent: 2929=M (agaagatgtctgagtacm) dbSNP|ss1765941, results in a Glycine at amino position 977 with either a C or A at that position. [0482]
  • SGP008 has a single nucleotide polymorphism at position 871:871=S (cagcagcctccgagggaaces). The accession number for this SNP in dbSNP is ss1389419. This is a non-silent change, with position 291 either a valine, when nucleotide 871=G, or a leucine, when nucleotide 871=C. This change could alter the biology of the enzyme. [0483]
  • Example 6
  • Isolation of cDNAs Encoding Mammalian Protein Phosphatases [0484]
  • Materials and Methods [0485]
  • Identification of Novel Clones [0486]
  • Total RNAs are isolated using the Guanidine Salts/Phenol extraction protocol of Chomczynski and Sacchi (P. Chomczynski and N. Sacchi, Anal. Biochem. 162, 156 (1987)) from primary human tumors, normal and tumor cell lines, normal human tissues, and sorted human hematopoietic cells. These RNAs are used to generate single-stranded cDNA using the Superscript Preamplification System (GIBCO BRL, Gaithersburg, Md.; Gerard, G F et al. (1989), [0487] FOCUS 11, 66) under conditions recommended by the manufacturer. A typical reaction uses 10 μg total RNA with 1.5 μg oligo(dT)12-18 in a reaction volume of 60 μL. The product is treated with RNaseH and diluted to 100 μL with H2O. For subsequent PCR amplification, 1-4 μL of this sscDNA is used in each reaction.
  • Degenerate oligonucleotides are synthesized on an Applied Biosystems 3948 DNA synthesizer using established phosphoramidite chemistry, precipitated with ethanol and used unpurified for PCR. These primers are derived from the sense and antisense strands of conserved motifs within the catalytic domain of several protein phosphatases. Degenerate nucleotide residue designations are: N=A, C, G, or T; R=A or G; Y═C or T; H=A, C or T not G; D=A, G or T not C; S═C or G; and W=A or T. [0488]
  • PCR reactions are performed using degenerate primers applied to multiple single-stranded cDNAs. The primers are added at a final concentration of 5 μM each to a mixture containing 10 mM TrisHCl, pH 8.3, 50 mM KCl, 1.5 mM MgCl[0489] 2, 200 μM each deoxynucleoside triphosphate, 0.001% gelatin, 1.5 U AmpliTaq DNA Polymerase (Perkin-Elmer/Cetus), and 1-4 μL cDNA. Following 3 min denaturation at 95° C., the cycling conditions are 94° C. for 30 s, 50° C. for 1 min, and 72° C. for 1 min 45 s for 35 cycles. PCR fragments migrating between 300-350 bp are isolated from 2% agarose gels using the GeneClean Kit (Bio101), and T-A cloned into the pCRII vector (Invitrogen Corp. U.S.A.) according to the manufacturer's protocol.
  • Colonies are selected for mini plasmid DNA-preparations using Qiagen columns and the plasmid DNA is sequenced using a cycle sequencing dye-terminator kit with AmpliTaq DNA Polymerase, FS (ABI, Foster City, Calif.). Sequencing reaction products are run on an ABI Prism 377 DNA Sequencer, and analyzed using the BLAST alignment algorithm (Altschul, S. F. et al., J. Mol. Biol. 215: 403-10). [0490]
  • Additional PCR strategies are employed to connect various PCR fragments or ESTs using exact or near exact oligonucleotide primers. PCR conditions are as described above except the annealing temperatures are calculated for each oligo pair using the formula: Tm=4(G+C)+2(A+T). [0491]
  • Isolation of cDNA Clones: [0492]
  • Human cDNA libraries are probed with PCR or EST fragments corresponding to phosphatase-related genes. Probes are [0493] 32P-labeled by random priming and used at 2×106 cpm/mL following standard techniques for library screening. Pre-hybridization (3 h) and hybridization (overnight) are conducted at 42° C. in 5×SSC, 5× Denhart's solution, 2.5% dextran sulfate, 50 mM Na2PO4/NaHPO4, pH 7.0, 50% formamide with 100 mg/mL denatured salmon sperm DNA. Stringent washes are performed at 65° C. in 0.1×SSC and 0.1% SDS. DNA sequencing is carried out on both strands using a cycle sequencing dye-terminator kit with AmpliTaq DNA Polymerase, FS (ABI, Foster City, Calif.). Sequencing reaction products are run on an ABI Prism 377 DNA Sequencer.
  • Example 7
  • Protein Phosphatase Gene Expression [0494]
  • Expression Vector Construction [0495]
  • Expression constructs are generated for some of the human cDNAs including: a) full-length clones in a pCDNA expression vector; b) a GST-fusion construct containing the catalytic domain of the novel phosphatase fused to the C-terminal end of a GST expression cassette; and c) a full-length clone containing a Cys to Ser (C to S) mutation at the predicted catalytic site within the phosphatase domain, inserted in the pCDNA vector. [0496]
  • The “C to S” mutants of the phosphatase might function as dominant negative constructs, and will be used to elucidate the function of these novel phosphatases. [0497]
  • Example 8
  • Generation of Specific Immunoreagents to Protein Phosphatases [0498]
  • Materials and Methods [0499]
  • Specific immunoreagents are raised in rabbits against KLH- or MAP-conjugated synthetic peptides corresponding to isolated phosphatase polypeptides. C-terminal peptides are conjugated to KLH with glutaraldehyde, leaving a free C-terminus. Internal peptides are MAP-conjugated with a blocked N-terminus. Additional immunoreagents can also be generated by immunizing rabbits with the bacterially expressed GST-fusion proteins containing the cytoplasmic domains of each novel PTP or STP. [0500]
  • The various immune sera are first tested for reactivity and selectivity to recombinant protein, prior to testing for endogenous sources. [0501]
  • Western Blots [0502]
  • Proteins in SDS PAGE are transferred to immobilon membrane. The washing buffer is PBST (standard phosphate-buffered saline pH 7.4+0.1% Triton X-100). Blocking and antibody incubation buffer is PBST +5% milk. Antibody dilutions varied from 1:1000 to 1:2000. [0503]
  • Example 9
  • Recombinant Expression and Biological Assays for Protein Phosphatases [0504]
  • Materials and Methods [0505]
  • Transient Expression of Phosphatases in Mammalian Cells [0506]
  • The pcDNA expression plasmids (10 μg DNA/100 mm plate) containing the STE20-related phosphatase constructs are introduced into 293 cells with lipofectamine (Gibco BRL). After 72 hours, the cells are harvested in 0.5 mL solubilization buffer (20 mM HEPES, pH 7.35, 150 mM NaCl, 10% glycerol, 1% Triton X-100, 1.5 mM MgCl[0507] 2, 1 mM EGTA, 2 mM phenylmethylsulfonyl fluoride, 1 μg/mL aprotinin). Sample aliquots are resolved by SDS polyacrylamide gel electrophoresis (PAGE) on 6% acrylamide/0.5% bis-acrylamide gels and electrophoretically transferred to nitrocellulose. Non-specific binding is blocked by preincubating blots in Blotto (phosphate buffered saline containing 5% w/v non-fat dried milk and 0.2% v/v Nonidet P-40 (Sigma)), and recombinant protein was detected using the various anti-peptide or anti-GST-fusion specific antisera.
  • In Vitro Phosphatase Assays [0508]
  • Three days after transfection with the phosphatase expression constructs, a 10 cm plate of 293 cells is washed with PBS and solubilized on ice with 2 mL PBSTDS containing phosphatase inhibitors (10 mM NaBPO[0509] 4, pH 7.25, 150 mM NaCl, 1% Triton X-100, 0.5% deoxycholate, 0.1% SDS, 0.2% sodium azide, 1 mM NaF, 1 mM EGTA, 4 mM sodium orthovanadate, 1% aprotinin, 5 μg/mL leupeptin). Cell debris is removed by centrifugation (12000×g, 15 min, 4° C.) and the lysate is precleared by two successive incubations with 50 μL of a 1:1 slurry of protein A sepharose for 1 hour each. One-half mL of the cleared supernatant is reacted with 10 μL of protein A purified phosphatase-specific antisera (generated from the GST fusion protein or antipeptide antisera) plus 50 μL of a 1:1 slurry of protein A-sepharose for 2 hr at 4° C. The beads are then washed 2 times in PBSTDS, and 2 times in HNTG (20 mM HEPES, pH 7.5/150 mM NaCl, 0.1% Triton X-100, 10% glycerol).
  • The immunopurified phosphatases on sepharose beads are resuspended in 20 μL HNTG plus 30 mM MgCl[0510] 2, 10 mM MnCl2, and 20 μCi [α32P]ATP (3000 Ci/mmol). The phosphatase reactions are run for 30 min at room temperature, and stopped by addition of HNTG supplemented with 50 mM EDTA. The samples are washed 6 times in HNTG, boiled 5 min in SDS sample buffer and analyzed by 6% SDS-PAGE followed by autoradiography. Phosphoamino acid analysis is performed by standard 2D methods on 32P-labeled bands excised from the SDS-PAGE gel.
  • Similar assays are performed on bacterially expressed GST-fusion constructs of the phosphatases. [0511]
  • Example 10
  • Demonstration of Gene Amplification by Southern Blotting [0512]
  • Materials and Methods [0513]
  • Nylon membranes are purchased from Boehringer Mannheim. Denaturing solution contains 0.4 M NaOH and 0.6 M NaCl. Neutralization solution contains 0.5 M Tris-HCL, pH 7.5 and 1.5 M NaCl. Hybridization solution contains 50% formamide, 6×SSPE, 2.5× Denhardt's solution, 0.2 mg/nl denatured salmon DNA, 0.1 mg/mL yeast tRNA, and 0.2% sodium dodecyl sulfate. Restriction enzymes are purchased from Boebringer Mannheim. Radiolabeled probes are prepared using the Prime-it II kit by Stratagene. The beta-actin DNA fragment used for a probe template is purchased from Clontech. [0514]
  • Genomic DNA is isolated from a variety of tumor cell lines (such as MCF-7, MDA-MB-231, Calu-6, A549, HCT-15, HT-29, Colo 205, LS-180, DLD-1, HCT-116, PC3, CAPAN-2, MIA-PaCa-2, PANC-1, AsPc-1, BxPC-3, OVCAR-3, SKOV3, SW 626 and PA-1, and from two normal cell lines. [0515]
  • A 10 μg aliquot of each genomic DNA sample is digested with EcoR I restriction enzyme and a separate 10 μg sample is digested with Hind III restriction enzyme. The restriction-digested DNA samples are loaded onto a 0.7% agarose gel and, following electrophoretic separation, the DNA is capillary-transferred to a nylon membrane by standard methods (Sambrook, J. et al (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory). [0516]
  • Example 11
  • Detection of Protein-Protein Interaction Through Phage Display [0517]
  • Materials And Methods [0518]
  • Phage display provides a method for isolating molecular interactions based on affinity for a desired bait. cDNA fragments cloned as fusions to phage coat proteins are displayed on the surface of the phage. Phage(s) interacting with a bait are enriched by affinity purification and the insert DNA from individual clones is analyzed. [0519]
  • T7 Phage Display Libraries [0520]
  • All libraries are constructed in the T7Select1-1b vector (Novagen) according to the manufacturer's directions. [0521]
  • Bait Presentation [0522]
  • Protein domains to be used as baits are generated as C-terminal fusions to GST and expressed in [0523] E. coli. Peptides are chemically synthesized and biotinylated at the N-terminus using a long chain spacer biotin reagent.
  • Selection [0524]
  • Aliquots of refreshed libraries (10[0525] 10-1012 pfu) supplemented with PanMix and a cocktail of E. coli inhibitors (Sigma P-8465) are incubated for 1-2 hrs at room temperature with the immobilized baits. Unbound phage is extensively washed (at least 4 times) with wash buffer.
  • After 3-4 rounds of selection, bound phage is eluted in 100 μL of 1% SDS and plated on agarose plates to obtain single plaques. [0526]
  • Identification of Insert DNAs [0527]
  • Individual plaques are picked into 25 μL of 10 mM EDTA and the phage is disrupted by heating at 70° C. for 10 min. 2 μL of the disrupted phage are added to 50 μL PCR reaction mix. The insert DNA is amplified by 35 rounds of thermal cycling (94° C., 50 sec; 50° C., 1 min; 72° C., 1 min). [0528]
  • Composition of Buffer [0529]
  • 10× PanMix [0530]
  • 5% Triton X-100 [0531]
  • 10% non-fat dry milk (Carnation) [0532]
  • 10 mM EGTA [0533]
  • 250 mM NaF [0534]
  • 250 μg/mL Heparin (sigma) [0535]
  • 250 μg/mL sheared, boiled salmon sperm DNA (sigma) [0536]
  • 0.05% Na azide [0537]
  • Prepared in PBS [0538]
  • Wash Buffer [0539]
  • PBS supplemented with: [0540]
  • 0.5% NP-40 [0541]
  • [0542]
  • [0543] 25 μg/mL heparin
  • PCR reaction mix [0544]
  • 1.0 [0545] mL 10× PCR buffer (Perkin-Elmer, with 15 mM Mg)
  • 0.2 mL each dNTPs (10 mM stock) [0546]
  • 0.1 mL T7UP primer (15 pmol/μL) GGAGCTGTCGTATTCCAGTC [0547]
  • 0.1 mL T7DN primer (15 pmol/μL) AACCCCTCAAGACCCGTTTAG [0548]
  • 0.2 mL 25 mM MgCl[0549] 2 or MgSO4 to compensate for EDTA
  • Q.S. to 10 mL with distilled water [0550]
  • Add 1 unit of Taq polymerase per 50 μL reaction [0551]
  • Library: T7 Select1-H441 [0552]
  • Compound Evaluation [0553]
  • It will be appreciated that, in any given series of compounds, a spectrum of biological activity will be observed. In a preferred embodiment, the present invention relates to compounds demonstrating the ability to modulate protein enzymes related to cellular signal transduction; preferably, protein phosphatases; and most preferably, protein tyrosine phosphatases. The assays described below are employed to select those compounds demonstrating the optimal degree of the desired activity. [0554]
  • As used herein, the phrase “optimal degree of desired activity” refers to the highest therapeutic index, defined above, against a protein enzyme which mediates cellular signal transduction and which is related to a particular disorder so as to provide an animal or a human patient, suffering from such disorder with a therapeutically effective amount of a compound of this invention at the lowest possible dosage. [0555]
  • Assays For Determining Inhibitory Activity [0556]
  • Various procedures known in the art may be used for identifying, evaluating or assaying the inhibition of activity of protein enzymes, in particular protein phosphatases, by the compounds of the invention. For example but without limitation, with regard to phosphatases such assays involve exposing target cells in culture to the compounds and (a) biochemically analyzing cell lysates to assess the level and/or identity of phosphorylated proteins; or (b) scoring phenotypic or functional changes in treated cells as compared to control cells that were not exposed to the test substance. [0557]
  • Where mimics of the natural ligand for a signal transducing receptor are to be identified or evaluated, the cells are exposed to the compound of the invention and compared to positive controls which are exposed only to the natural ligand, and to negative controls which are not exposed to either the compound or the natural ligand. For receptors that are known to be phosphorylated at a basal level in the absence of the natural ligand, such as the insulin receptor, the assay may be carried out in the absence of the ligand. Where inhibitors or enhancers of ligand induced signal transduction are to be identified or evaluated, the cells are exposed to the compound of the invention in the presence of the natural ligand and compared to controls which are not exposed to the compound of the invention. [0558]
  • The assays described below may be used as a primary screen to evaluate the ability of the compounds of this invention to inhibit phosphatase activity of the compounds of the invention. The assays may also be used to assess the relative potency of a compound by testing a range of concentrations, in a range from 100 μM to 1 pM, for example, and computing the concentration at which the amount of phosphorylation or signal transduction is reduced or increased by 50% (IC50) compared to controls. [0559]
  • Biochemical Assays [0560]
  • In one embodiment target cells having a substrate molecule that is phosphorylated or dephosphorylated on a tyrosine residue during signal transduction are exposed to the compounds of the invention and radiolabelled phosphate, and thereafter, lysed to release cellular contents, including the substrate of interest. The substrate may be analyzed by separating the protein components of the cell lysate using a sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of phosphorylated proteins by exposing to X-ray film. In a similar technique, but without radioactive labeling, the protein components separated by SDS-PAGE are transferred to a nitrocellulose membrane, the presence of pTyr is detected using an antiphosphotyrosine (anti-pTyr) antibody. Alternatively, it is preferred that the substrate of interest be first isolated by incubating the cell lysate with a substrate-specific anchoring antibody bound to a solid support, and thereafter, washing away non-bound cellular components, and assessing the presence or absence of pTyr on the solid support by an anti-pTyr antibody. This preferred method can readily be performed in a microtiter plate format by an automated robotic system, allowing for testing of large numbers of samples within a reasonably short time frame. [0561]
  • The anti-pTyr antibody can be detected by labeling it with a radioactive substance which facilitates its detection by autoradiography. Alternatively, the anti-pTyr antibody can be conjugated with an enzyme, such as horseradish peroxidase, and detected by subsequent addition of an appropriate substrate for the enzyme, the choice of which would be clear to one skilled in the art. A further alternative involves detecting the anti-pTyr antibody by reacting with a second antibody which recognizes the anti-pTyr antibody, this second antibody being labeled with either a radioactive substance or an enzyme as previously described. Any other methods for the detection of an antibody known in the art may be used. [0562]
  • The above methods may also be used in a cell-free system wherein cell lysate containing the signal-transducing substrate molecule and phosphatase is mixed with a compound of the invention and a kinase. The substrate is phosphorylated by initiating the kinase reaction by the addition of adenosine triphosphate (ATP). To assess the activity of the compound, the reaction mixture may be analyzed by the SDS-PAGE technique or it may be added to a substrate-specific anchoring antibody bound to a solid support, and a detection procedure as described above is performed on the separated or captured substrate to assess the presence or absence of pTyr. The results are compared to those obtained with reaction mixtures to which the compound is not added. The cell-free system does not require the natural ligand or knowledge of its identity. For example, Posner et al. (U.S. Pat. No. 5,155,031) describes the use of insulin receptor as a substrate and rat adipocytes as target cells to demonstrate the ability of pervanadate to inhibit PTP activity. Burke et al., 1994, [0563] Biochem. Biophys. Res. Comm., 204:129-134) describes the use of autophosphorylated insulin receptor and recombinant PTP1B in assessing the inhibitory activity of a phosphotyrosyl mimetic.
  • In addition to measuring phosphorylation or dephosphorylation of substrate proteins, activation or modulation of second messenger production, changes in cellular ion levels, association, dissociation or translocation of signaling molecules, gene induction or transcription or translation of specific genes may also be monitored. These biochemical assays may be performed using conventional techniques developed for these purposes. [0564]
  • Biological Assays [0565]
  • The ability of the compounds of this invention to modulate the activity of PTPs, which control signal transduction, may also be measured by scoring for morphological or functional changes associated with ligand binding. Any qualitative or quantitative techniques known in the art may be applied for observing and measuring cellular processes which come under the control of phosphatases in a signaling pathway. Such cellular processes may include, but are not limited to, anabolic and catabolic processes, cell proliferation, cell differentiation, cell adhesion, cell migration and cell death. [0566]
  • The techniques that have been used for investigating the various biological effects of vanadate as a phosphatase inhibitor may be adapted for use with the compounds of the invention. For example, vanadate has been shown to activate an insulin-sensitive facilitated transport system for glucose and glucose analogs in rat adipocytes (Dubyak et al., 1980, [0567] J. Biol. Chem., 256:5306-5312). The activity of the compounds of the invention may be assessed by measuring the increase in the rate of transport of glucose analog such as 2-deoxy-3H-glucose in rat adipocytes that have been exposed to the compounds. Vanadate also mimics the effect of insulin on glucose oxidation in rat adipocytes (Shechter et al., 1980, Nature, 284:556-558). The compounds of this invention may be tested for stimulation of glucose oxidation by measuring the conversion of 14C-glucose to 14CO2. Moreover, the effect of sodium orthovanadate on erythropoietin-mediated cell proliferation has been measured by cell cycle analysis based on DNA content as estimated by incorporation of tritiated thymidine during DNA synthesis (Spivak et al., 1992, Exp. Hematol., 20:500-504). Likewise, the activity of the compounds of this invention toward phosphatases that play a role in cell proliferation may be assessed by cell cycle analysis.
  • The activity of the compounds of this invention can also be assessed in animals using experimental models of disorders caused by or related to dysfunctional signal transduction. For example, the activity of a compound of this invention may be tested for its effect on insulin receptor signal transduction in non-obese diabetic mice (Lund et a., 1990, [0568] Nature, 345:727-729), B B Wistar rats and streptozotocin-induced diabetic rats (Solomon et al., 1989, Am. J Med. Sci., 297:372-376). The activity of the compounds may also be assessed in animal carcinogenesis experiments since phosphatases can play an important role in dysfunctional signal transduction leading to cellular transformation. For example, okadaic acid, a phosphatase inhibitor, has been shown to promote tumor formation on mouse skin (Suganuma et al., 1988, Proc. Natl. Acad. Sci., 85:1768-1771).
  • The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages for use in humans. The dosage of the compounds of the invention should lie within a range of circulating concentrations with little or no toxicity. The dosage may vary within this range depending on the dosage form employed and the route of administration. [0569]
  • Phosphotyrosine Enzyme Linked Immunosorbent Assay [0570]
  • This assay may be used to test the ability of the compounds of the invention to inhibit dephosphorylation of phosphotyrosine (pTyr) residues on insulin receptor (IR). Those skilled in the art will recognize that other substrate molecules, such as platelet derived growth factor receptor, may be used in the assay by using a different target cell and anchoring antibody. By using different substrate molecules in the assay, the activities of the compounds of this invention toward different protein tyrosine enzymes may be assessed. In the case of IR, an endogenous kinase activity is active at low level even in the absence of insulin binding. Thus, no insulin is needed to stimulate phosphorylation of IR. That is, after exposure to a compound, cell lysates can be prepared and added to microtiter plates coated with anti-insulin receptor antibody. The level of phosphorylation of the captured insulin receptor is detected using an anti-pTyr antibody and an enzyme-linked secondary antibody. [0571]
  • Assay Methods in Determination of Compound-PTP IC50 [0572]
  • The following in vitro assay procedure is preferred to determine the level of activity and effect of the different compounds of the present invention on one or more of the PTPs. Similar assays can be designed along the same lines for any PTP using techniques well known in the art. [0573]
  • The catalytic assays described herein are performed in a 96-well format. The general procedure begins with the determination of PTP optimal pH using a three-component buffer system that minimizes ionic strength variations across a wide range of buffer pH. Next, the Michaelis-Menten constant, or Km, is determined for each specific substrate-PTP system. This Km value is subsequently used as the substrate reaction concentration for compound screening. Finally, the test PTP is exposed to varying concentrations of compound for fifteen minutes and allowed to react with substrate for ten minutes. The results are plotted as percent inhibition versus compound concentration and the IC50 interpolated from the plot. [0574]
  • The following materials and reagents are used: [0575]
  • 1. Assay Buffer is used as solvent for all assay solutions unless otherwise indicated. [0576]
    Component Concentration
    Acetate (Fisher Scientific A38-500) 100 mM
    Bis-Tris (Sigma B-7535)  50 mM
    Tri-3 s (Fisher Scientific BP152-5)  50 mm
    Glycerol (Fisher Scientific BP229-1)  10% (v/v)
  • *1 mM DTT is added immediately prior to use [0577]
  • 2. 96 Well Easy Wash Plate (Costar 3369) [0578]
  • 3. p-Nitrophenyl Phosphate (Boehringer Mannheim 738−379) [0579]
  • 4. Fluorescein Diphosphate (Molecular Probes F-2999) [0580]
  • 5. 0.22 μm Stericup Filtration System 500 ml (Millipore SCGPU05RE) [0581]
  • 6. 10N NaOH (Fisher Scientific SS255-1) [0582]
  • 7. 10N HCl Fisher Scientific A144-500) [0583]
  • 8. Compounds were dissolved in DMSO (Sigma D-5879) at 5 or 10 mM concentrations and stored at −20° C. in small aliquots. [0584]
  • Methods: [0585]
  • All assays are performed using pNPP or FDP as substrate. The optimum pH is determined for each PTP used. [0586]
  • PTP Assay [0587]
  • PTPase activity is assayed at 25° C. in a 100-μl reaction mixture containing an appropriate concentration of pNPP or FDP as substrate. The reaction is initiated by addition of the PTP and quenched after 10 min by addition of 50 μl of 1N NaOH. The non-enzymatic hydrolysis of the substrate is corrected by measuring the control without the addition of the enzyme. The amount of p-nitrophenol produced is determined from the absorbance at 410 mn. To determine the kinetic parameter, Km, the initial velocities are measured at various substrate concentrations and the data are fitted to the Michaelis equation where velocity=(Vmax*[S])/(Km+[S]), and [S]=substrate reaction concentration. [0588]
  • Inhibition Studies [0589]
  • The effect of the compounds on PTP is evaluated at 25° C. using pNPP or FDP as substrate. PTP is pre-incubated for fifteen minutes with various concentrations of compound. Substrate is then added at a fixed concentration (usually equal to the Km previously calculated). After 10 minutes, NaOH is added to stop the reaction. The hydrolysis of pNPP is followed at 410 nm on the Biotek Powerwave 200 microplate scanning spectrophotometer. The percent inhibition is calculated as follows: Percent Inhibition=[(control signal−compound signal)/control signal]×100%. The IC50 is then determined by interpolation of a percent inhibition versus compound concentration plot. [0590]
  • Plasmids designed for bacterial GST-PTP fusion protein expression are derived by insertion of PCR-generated human PTP fragments into pGEX vectors (Pharmacia Biotech). Several of these constructs are then used to subclone phosphatases into pFastBac-1 for expression in Sf-9 insect cells. Oligonucleotides that are used for the initial amplification of PTP genes are shown below. The cDNAs are prepared using the Gilbo BRL superscript preamplification system on RNAs purchased from Clontech. [0591]
  • Conclusion [0592]
  • One skilled in the art would readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The molecular complexes and the methods, procedures, treatments, molecules, specific compounds described herein are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. It will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. [0593]
  • All patents and publications mentioned in the specification are indicative of the levels of those skilled in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference. [0594]
  • The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. Thus, for example, in each instance herein any of the terms “comprising,” “consisting essentially of” and “consisting of” may be replaced with either of the other two terms. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims. [0595]
  • In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group. For example, if X is described as selected from the group consisting of bromine, chlorine, and iodine, claims for X being bromine and claims for X being bromine and chlorine are fully described. [0596]
  • In view of the degeneracy of the genetic code, other combinations of nucleic acids also encode the claimed peptides and proteins of the invention. For example, all four nucleic acid sequences GCT, GCC, GCA, and GCG encode the amino acid alanine. Therefore, if for an amino acid there exists an average of three codons, a polypeptide of 100 amino acids in length will, on average, be encoded by 3100, or 5×1047, nucleic acid sequences. Thus, a nucleic acid sequence can be modified to form a second nucleic acid sequence, encoding the same polypeptide as encoded by the first nucleic acid sequences, using routine procedures and without undue experimentation. Thus, all possible nucleic acids that encode the claimed peptides and proteins are also fully described herein, as if all were written out in full taking into account the codon usage, especially that preferred in humans. Furthermore, changes in the amino acid sequences of polypeptides, or in the corresponding nucleic acid sequence encoding such polypeptide, may be designed or selected to take place in an area of the sequence where the significant activity of the polypeptide remains unchanged. For example, an amino acid change may take place within a β-turn, away from the active site of the polypeptide. Also changes such as deletions (e.g. removal of a segment of the polypeptide, or in the corresponding nucleic acid sequence encoding such polypeptide, which does not affect the active site) and additions (e.g. addition of more amino acids to the polypeptide sequence without affecting the function of the active site, such as the formation of GST-fusion proteins, or additions in the corresponding nucleic acid sequence encoding such polypeptide without affecting the function of the active site) are also within the scope of the present invention. Such changes to the polypeptides can be performed by those with ordinary skill in the art using routine procedures and without undue experimentation. Thus, all possible nucleic and/or amino acid sequences that can readily be determined not to affect a significant activity of the peptide or protein of the invention are also fully described herein. [0597]
  • The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein. [0598]
  • Other embodiments are within the following claims. [0599]
  • 1 76 1 6374 DNA Homo sapiens 1 acgtctgtgg cgccctcgca ccgcgccgca gccatggccc tggtgaccct gcagcgctcg 60 cccacgccca gcgccgcctc ctcctcggcc agcaacagcg agttggaggc tggcagcgaa 120 gaagatcgaa aattaaacct cagcttaagt gagagctttt tcatggtgaa aggcgcagcc 180 ctcttcttac aacagggaag cagccctcaa ggccagcgga gtcttcagca cccccacaag 240 catgcaggtg atctgcctca acatcttcag gtgatgatca accttctgcg ttgcgaagac 300 agaatcaagc tggcagtgcg cctggagagc gcctgggcgg accgggtccg gtacatggtg 360 gtggtgtaca gcagcgggcg ccaggacacc gaggagaata tcttgctggg agtggacttt 420 tccagtaagg aaagtaaaag ctgcaccatt gggatggttc tccgactgtg gagcgacacg 480 aaaatccacc ttgatggaga tggtgggttc agcgtgagca cagcaggaag gatgcacata 540 tttaagcctg tgtctgtcca ggccatgtgg tctgccctgc aggtgcttca caaggcctgc 600 gaagtggccc ggaggcacaa ctacttcccc gggggtgtag ctctcatctg ggctacctac 660 tatgagagct gcatcagctc cgagcagagc tgcatcaacg agtggaacgc catgcaggac 720 ctggagtcta cgcggcccga ctcccccgcg ctatttgtgg acaagcccac tgaaggggaa 780 aggaccgagc gcctcatcaa agccaagctc cgaagcatca tgatgagcca ggatctagaa 840 aatgtgactt ccaaagagat tcgtaatgaa ttagagaaac agatgaattg taacttgaag 900 gaactcaagg aatttataga caatgagatg ctacttatct tgggacagat ggacaagccc 960 tcccttatct tcgatcatct ttatctcggc tctgaatgga atgcatccaa tctggaggaa 1020 ctgcagggct caggggttga ttacatttta aatgttacca gagaaatcga taattttttt 1080 cctggcttat ttgcatatca taacatccga gtctacgatg aagagaccac agacctcctc 1140 gcccactgga atgaagcgta tcattttata aacaaagcga agaggaacca ttccaagtgc 1200 ctggtgcatt gcaaaatggg cgtgagtcgc tcggcctcca cagtcatagc ctatgcaatg 1260 aaggaattcg gctggcctct ggaaaaagca tataactatg taaagcagaa gcgcagcatc 1320 acgcgcccca acgcgggctt tatgaggcag ctgtctgagt atgaaggcat cttggatgca 1380 agcaaacagc ggcacaacaa gctgtggcgt cagcagacag acagcagcct ccagcagcct 1440 gtggatgacc ctgcaggacc tggcgacttc ttgccagaga ccccagatgg caccccggaa 1500 agccagctgc ccttcttgga tgatgccgcc cagcccggct tagggccccc cctcccctgc 1560 tgtttccggc gactctcaga cccccttctg ccttcccctg aggatgaaac tggcagcttg 1620 gtccacctgg aggatccgga gagggaggct ctgttggagg aagctgctcc acctgcagag 1680 gtgcacaggc cggccagaca gccccagcaa ggttccggac tctgtgagaa ggatgtgaag 1740 aagaaactag agtttgggag tcccaaaggt cggagcggct ccttgctgca ggtggaggag 1800 acggaaaggg aggagggcct gggagcaggg aggtgggggc agcttccaac ccagctcgat 1860 caaaacctgc tcaactcgga gaacctaaac aacaacagca agaggagctg tcccaacggc 1920 atggaggatg atgctatatt tgggatcctt aacaaagtga agccttccta taaatcctgt 1980 gccgactgca tgtaccctac agccagcggg gctcctgagg cctccaggga gcgatgtgag 2040 gaccccaatg ctcccgccat ctgcacccag ccagccttcc taccccacat cacgtcctcc 2100 cctgtggccc acttggccag caggtcccgt gttccggaga agccagcctc tggcccaacc 2160 gaacctcccc cgttcctacc accagcaggc tccaggaggg cagacaccag tggccctggg 2220 gctggagctg ccctagagcc accagccagc cttttggaac cttccagaga gaccccaaaa 2280 gtcctgccaa agtccctcct tttgaagaat tctcactgtg ataagaaccc tcccagcaca 2340 gaagtggtaa taaaggaaga atcgtcaccc aagaaagata tgaagccagc caaggacctg 2400 aggcttctgt tcagtaatga atctgagaag ccgacaacca acagctacct gatgcagcac 2460 caggagtcca tcattcagct gcagaaggca ggcttggtcc gcaagcacac caaagagcta 2520 gagcggctga agagcgtgcc tgcagaccca gcacctccct ccagggatgg ccctgccagc 2580 aggctggagg ccagcatccc cgaggagagc caggatccag ccgcgctcca cgagctgggc 2640 cccctggtta tgcccagcca ggccgggagt gatgagaagt cagaggccgc ccccgcttca 2700 ttggaaggag gctcactgaa gagcccccct cctttcttct accgcctgga ccacaccagt 2760 agtttctcaa aagactttct gaagaccatc tgctacaccc ccacctcctc ttccatgagc 2820 tccaacctga cccggagctc cagcagcgat agcatccaca gtgtccgtgg gaagcccggg 2880 ctggtgaagc agcggacaca ggagattgag acccggctcc ggctggcggg cctcaccgtc 2940 tcttccccac tgaagcgctc acactctctt gccaagctgg ggagtctcac cttctcaacg 3000 gaagacctgt ccagtgaggc tgacccgtcc accgtcgctg actcccagga caccactttg 3060 agtgaatctt ccttcttgca tgagccccag ggaaccccga gggacccagc tgcaacctcc 3120 aaaccatcag ggaaacccgc cccagaaaac ttaaagagcc cttcgtggat gagcaaaagc 3180 tgacccgcct tttgctgcgt caggctgggc ggatattttc atatgatccc tccctcactt 3240 tcactgtgga tttggatcga ccccttacat cttgatttcc cttaaacgag tgaagtcatc 3300 ctaacttttc ccctactctt gccatagaga ggaggagaag aaactaacca acatgcagca 3360 caagaagggg agctgtcagt cgtgtggcct gggagaacca ggagccgccg gccaggagac 3420 acaaaactcc gctcccaccg tgtcttcaag aagaaccatg ttttaggaag aacgtgcaca 3480 cataggcgca cacatccaga ctgttccctt cgcatcctgc agaagagtcc tggtggtggc 3540 agcctcaccg cggcacacgt tctagctcat tcttggcctc gcagaaaact ctcggatggc 3600 aacattaagt cctacctctg ttctcgctgc gctttttatt tttaaaacac acgaatgacc 3660 aaggcttgtt ccagggaata atgctctgtc tgaaagcgtt tttccaaaaa aagaattttt 3720 cgttttgtgt tttggttggg accccagacg tgtcaagact tttaacccga cagccccaag 3780 cactgctgag taaagtcatc agatgagtag tcccacgctg ttgttgagct gactctgtgc 3840 ccgaatggtg gtgcaggcgc cctgtcctgg ttgtgttgtt acgtgtttga ccagcacagg 3900 ggtccggtgg ggaaaatgta tgggttgttc tcagttgttg ctaatgctga agtttaaatc 3960 tcaaggggaa gggcccacct gcattgttga gtgtctgctg ctgaaacaca tgattgtgtt 4020 taggtttgaa attgctcaag tgtctggctc aggtggtggt tctgagacac atcgtcctgc 4080 tgagagccca gatgcttagg tccactaggg cccatctagg gaagggaaag gagatttcag 4140 cggcttcccc gaaaggaacg ggactgtcgg gatgcttccc ggatgtctac agttgcccct 4200 tcctgcagtg agattactgc ttcctgtttc cctccagctc ttcccagcag cagtgaggga 4260 gtataagagg gatctgtagt cgctgcctgg ctctgtgggc ggccccttta agactcaggt 4320 gagctcagcc agtcccgctg cgccaggctt gaatcaaagg tgtcagcaag ctagatgtca 4380 aaacatttcg agagagaggc agcttgcaga aaagcagaaa ggttgaattc aggggtcagc 4440 aaactatggc atgtggcccc gtgagctagg aatggttttt cctctttaag acagggcctg 4500 gctctgtcac ctaggctgga gtacagtagt gcaatcatga ctcactgcag catcaacctc 4560 cctggctcaa gcgatggctt ttaccttttt aaggagttgt aaaaaaaaga aataatatgt 4620 gacagaattc atgtggccca caaagcctga ggtacttact gtcaggccct ttgtagaaaa 4680 agttgctgac ccctggtttg atttgccaca cagggctgct catggccctg taggaagaaa 4740 ctgggtaaat gtgaagccct ctgcctctct gacacctaag gccagaccct tccctcctcc 4800 cgaccacaag ctttgccagc cccgcactag cctcacctgt gcaggatgga gtaggttgcg 4860 tacatggggg aaggcaggtg aacacagttg gctgaagctc ttccacaatt ccatcttgcc 4920 ctcagctggg tccgccagat taactcagtg aaaccagaaa gccttcaagg accagctgaa 4980 ttctgaaagt gagtgagtga gccatcatct taacattggc cagaactgtg ttccccaaag 5040 ctattctaga aagcacccca ggagggatct gcaggaacaa ggctagttca tattttacct 5100 agtgagcaca gtttttgcaa aaatcctccc tccaggactt tgtctccttg gagtgatttt 5160 taaaaataca tacattactt tatagggagc tgttttccca ctagtgtcaa ttaaaatcac 5220 cttaaaaggt gattatccac ttattcctaa acccctgtgg gttgttcccc ctttccctca 5280 gccaacaaaa gcatagcctc aaaaaatatc aagttcggta tgttttgcca aatcaaattt 5340 catgtggtag atcaattttt gtgtcaaaat aatcttttaa atttagtgat gacaggcttt 5400 tgttggtttt ttaaccacgt ctatgtatga gaatgatata tttttgaaaa ctttaatttt 5460 gaaagccata atttttctta tctaaagagt tggggggtgg ggtgtggaat ctggagagta 5520 caagttggtc tttggcttct ggcaaactta cccattcatt tttggaagca cagctagcat 5580 atcaacatcc agacgagagc gctggtcccg tccacagagc agagtgaagc attctggact 5640 tgatgcttaa tagcctggcc tggagaaaag ggtaaggttt atttttggaa acccagatca 5700 gttgcatgta aacagatggc acatggctat ttaaaatgct gtatgatggg gccgggcgcg 5760 gtggctcacg cctgtaatcc tagcactttg ggaggccgag gcaggcggat catctgaggt 5820 caggagttct agaccagcct ggccaacatg gtgaaacccc gtctctacta aaaatacaaa 5880 aacttagctg ggtgtggtag tgcgtgcctg taatcctagc tacttaggag gctgaggcag 5940 gagaatcgct tgaacctggg tggcggaggt tgcaatgagc cgagatcgcg ccactgcact 6000 ccagcctggg tgcaatgaaa ctgggagtga aactccgtct caaaaaaata aaatgctgta 6060 tgaacaagat gagcattctg tcaggtgtcg ggacacctgg gcaaagacga attcatgctg 6120 tctgtgaaaa ggaagtttgc actgtaacat atgccatagc ttggcccttg ctttgtatgc 6180 aaccttagct gatggggaaa attccaaaca taagtggcac aggaaagagg caggcagagg 6240 aggcaggggt ctgtgctgtg caagtcactg gttttttatg actatcattt tcattgaatg 6300 catttgttga attgggacaa aaggaacatt ttctaaatca gcttgatact ctttataaaa 6360 aacagctgaa tctt 6374 2 2732 DNA Homo sapiens 2 gaaaagaaga cgaggaggag agcgacggga cgggacgcga gcgggagcgc agccgccctc 60 tcggctccgc ggcggcgcct cgcaagtccg ggaggcgagg ggggcccgag gggagacgcc 120 gtgacaactt tcgtttccct ctgagggaat tgggaggtcg gcggccccaa aagctttcag 180 tccagtgtaa agctgttgga gcgcgggagc aaaggtaaag aatgatgtaa tgcgctggct 240 gctccaaagc atcttttgtt gtggaatggt tattccagtc atctctttat gaatcaaatg 300 tgaggggctg ctttgtggac ggagtccttt gcaagagcac atcaacggga aagagaaaga 360 gacattcact tggagggctc ttgctgaaaa tgggtttaac tctccttttg ccagtcacca 420 ccagcctgac ctcatacact tttagtacaa tggagtggct gagcctttga gcacaccacc 480 attacatcat cgtggcaaat taaagaagga ggtgggaaaa gaggacttat tgttgtcatg 540 gcccatgaga tgattggaac tcaaattgtt actgagaggt tggtggctct gctggaaagt 600 ggaacggaaa aagtgctgct aattgatagc cggccatttg tggaatacaa tacatcccac 660 attttggaag ccattaatat caactgctcc aagcttatga agcgaaggtt gcaacaggac 720 aaagtgttaa ttacagagct catccagcat tcagcgaaac ataaggttga cattgattgc 780 agtcagaagg ttgtagttta cgatcaaagc tcccaagatg ttgcctctct ctcttcagac 840 tgttttctca ctgtacttct gggtaaactg gagaagagct tcaactctgt tcacctgctt 900 gcaggtgggt ttgctgagtt ctctcgttgt ttccctggcc tctgtgaagg aaaatccact 960 ctagtcccta cctgcatttc tcagccttgc ttacctgttg ccaacattgg gccaacccga 1020 attcttccca atctttatct tggctgccag cgagatgtcc tcaacaagga gctgatgcag 1080 cagaatggga ttggttatgt gttaaatgcc agcaatacct gtccaaagcc tgactttatc 1140 cccgagtctc atttcctgcg tgtgcctgtg aatgacagct tttgtgagaa aattttgccg 1200 tggttggaca aatcagtaga tttcattgag aaagcaaaag cctccaatgg atgtgttcta 1260 gtgcactgtt tagctgggat ctcccgctcc gccaccatcg ctatcgccta catcatgaag 1320 aggatggaca tgtctttaga tgaagcttac agatttgtga aagaaaaaag acctactata 1380 tctccaaact tcaattttct gggccaactc ctggactatg agaagaagat taagaaccag 1440 actggagcat cagggccaaa gagcaaactc aagctgctgc acctggagaa gccaaatgaa 1500 cctgtccctg ctgtctcaga gggtggacag aaaagcgaga cgcccctcag tccaccctgt 1560 gccgactctg ctacctcaga ggcagcagga caaaggcccg tgcatcccgc cagcgtgccc 1620 agcgtgccca gcgtgcagcc gtcgctgtta gaggacagcc cgctggtaca ggcgctcagt 1680 gggctgcacc tgtccgcaga caggctggaa gacagcaata agctcaagcg ttccttctct 1740 ctggatatca aatcagtttc atattcagcc agcatggcag catccttaca tggcttctcc 1800 tcatcagaag atgctttgga atactacaaa ccttccacta ctctggatgg gaccaacaag 1860 ctatgccagt tctcccctgt tcaggaacta tcggagcaga ctcccgaaac cagtcctgat 1920 aaggaggaag ccagcatccc caagaagctg cagactgcca ggccttcaga cagccagagc 1980 aagcgattgc attcggtcag aaccagcagc agtggcaccg cccagaggtc ccttttatct 2040 ccactgcatc gaagtgggag cgtggaggac aattaccaca ccagcttcct tttcggcctt 2100 tccaccagcc agcagcacct cacgaagtct gctggcctgg gccttaaggg ctggcactcg 2160 gatatcttgg ccccccagac ctctacccct tccctgacca gcagctggta ttttgccaca 2220 gagtcctcac acttctactc tgcctcagcc atctacggag gcagtgccag ttactctgcc 2280 tacagctgca gccagctgcc cacttgcgga gaccaagtct attctgtgcg caggcggcag 2340 aagccaagtg acagagctga ctcgcggcgg agctggcatg aagagagccc ctttgaaaag 2400 cagtttaaac gcagaagctg ccaaatggaa tttggagaga gcatcatgtc agagaacagg 2460 tcacgggaag agctggggaa agtgggcagt cagtctagct tttcgggcag catggaaatc 2520 attgaggtct cctgagaaga aagacacttg tgacttctat agacaatttt tttttcttgt 2580 tcacaaaaaa attccctgta aatctgaaat atatatatgt acatacatat atatttttgg 2640 aaaatggagc tatggtgtaa aagcaacagg tggatcaacc cagttgttac tctcttaaca 2700 tctgcatttg agagatcagc taatacttct ct 2732 3 2260 DNA Homo sapiens 3 ggggggaaaa gttaagaaaa agcccccgag agccggggtg aagggagtaa actggtctag 60 cccagttctg tctgcgccca gtgagagggt ttgaaactcc gcggagccct ttcccaatag 120 aaaacgtgtt tgcttcagga ttttcatctc agctgctttt ttttaaggtt ggttaagctg 180 tgcgccgccc cacctcataa aatggttgtc tctcaggagt gaagactcca gcggcccccc 240 gccctgggac ccctcaccgg gctggctccc ttcggcccct tcccccaccc cactccacag 300 cacccagtgc tggcaaaccg cgcgattcca gcacgaagga ggaaacccag gagggtgcgg 360 cggcccgagg cgcacgcact cggccagctt ccggcaactc aagggttacg accaggcggc 420 ggcgcgcgcc gaggggagag gcggtagctg acaggtggcg cctgcgcact gggagcgctc 480 attgtgcccc gcagctgccg aaccgcccgc ccgccgcccg ttcggagcgt cagtcggcga 540 cagtctggtg gtgggtgcgg agtctgcggc cgttcccgcg gcctcctcct cctccccgtt 600 cccttcaccc ccaccccgca cccctttccc catcccggct ccgtcaccct cccgtccccc 660 acactcagga caagaatgcc ctgcccggaa caacccagca gcgcctagat ggctttggtc 720 acggtccagc ggtcacctac ccccagcacc acctccagcc cctgcgcctc ggaggcagac 780 agtggggagg aagaatgccg gtcacagccc aggagcatca gcgagagctt tctaactgtc 840 aaaggtgctg ccctttttct accacgggga aatggctcat ccacaccaag aatcagccac 900 agacggaaca agcatgcagg cgatctccaa cagcatctcc aagcaatgtt cattttactc 960 cgcccagaag acaacatcag gctggctgta agactggaaa gtacttacca gaatcgaaca 1020 cgctatatgg tagtggtttc aactaatggt agacaagaca ctgaagaaag catcgtccta 1080 ggaatggatt tctcctctaa tgacagtagc acttgtacca tgggcttagt tttgcctctc 1140 tggagcgaca cgctaattca tttggatggt gatggtgggt tcagtgtatc gacggataac 1200 agagttcaca tattcaaacc tgtatctgtg caggcaatgt ggtctgcact acagagctta 1260 cacaaggctt gtgaagtcgc cagagcgcat aactactacc caggcagcct atttctcact 1320 tgggtgagtt attatgagag ccatatcaac tcagatcaat cctcagtcaa tgaatggaat 1380 gcaatgcaag atgtacagtc ccaccggccc gactctccag ctctcttcac cgacatacct 1440 actgaacgtg aacgaacaga aaggctaatt aaaaccaaat taagggagat catgatgcag 1500 aaggatttgg agaatattac atccaaagag ataagaacag agttggaaat gcaaatggtg 1560 tgcaacttgc gggaattcaa ggaatttata gacaatgaaa tgatagtgat ccttggtcaa 1620 atggatagcc ctacacagat atttgagcat gtgttcctgg gctcagaatg gaatgcctcc 1680 aacttagagg acttacagaa ccgaggggta cggtatatct tgaatgtcac tcgagagata 1740 gataacttct tcccaggagt ctttgagtat cataacattc gggtatatga tgaagaggca 1800 acggatctcc tggcgtactg gaatgacact tacaaattca tctctaaagc aaagaaacat 1860 ggatctaaat gccttgtgca ctgcaaaatg ggggtgagtc gctcagcctc caccgtgatt 1920 gcctatgcaa tgaaggaata tgaccgagcc tatgactatg tgaaagaaag acgaacggta 1980 accaagccca acccaagctt catgagacaa ctggaagagt atcaggggat cttgctggca 2040 agcttcctag gcttgattca tggagggagg gacaagccct ggggagagaa aagcacagaa 2100 tttgagtcag tagatctggt ttccattcct ggttcaccct cttgctgcaa ccctgagaag 2160 ttacttcaca tttctcatcc ttacctgacc ccatctataa aatgaaaatc aagagatcca 2220 tctcacaggg ttattgtgaa taaaaatgtg tttgaatgtt 2260 4 4360 DNA Homo sapiens 4 gaagaagccc tgaggaagga ggtggctggt tcctgtctca ctggccggag cttcagcttc 60 agtcatttca tctgggtccc tcagcccttg gtggggaaca tccaggcagg ttagaggtga 120 gtgtgcttct ccccttgtca atcctggtga gcccaggact tacaatacaa ggggcagcgg 180 ctttgccctg agtgtcaggg cacaaccatg atggctggga ccagctgctg gtatccttca 240 tgtcctttaa taggctccag gatgacgcct gagccaaagg ccctacctcc tgtggccttg 300 gttagagaca ccgaaggcca gctgtgtctt ccccagcaga gacaaagagg ttggcaggtt 360 gtcatggcga ccagaaagga cacagaggag gagcaggtag tcccaagcga ggaggacgaa 420 gccaacgtga gggcggtgca ggcccactac ctccgaagcc cctcccctag ccagtattcg 480 atggtctcag atgcagaaac agaaagcatt ttcatggaac ccattcacct ctcctcagcc 540 attgcagcca aacagatcat caatgaagaa ctcaagccac cgggggtcag agcagacgca 600 gagtgtccag gcatgctgga gtctgctgaa cagctgctgg tggaggacct gtacaaccgc 660 gtcagggaga agatggatga caccagcctc tataatacgc cctgtgtcct ggacctacag 720 cgggccctgg ttcaggatcg ccaagaggcg ccctggaatg aggtggatga ggtctggccc 780 aatgtcttca tagctgagaa gagtgtggct gtgaacaagg ggaggctgaa gaggctggga 840 atcacccaca ttctgaatgc tgcgcatggc accggcgttt acactggccc cgaattctac 900 actggcctgg agatccagta cctgggtgta gaggtggatg actttcctga ggtggacatt 960 tcccagcatt tccggaaggc gtactgtcat tacatcattt tctcttgtgt tttcatttca 1020 gggaaagtcc tggtcagcag cgaaatgggc atcagccggt cagcagtgct ggtggtcgcc 1080 tacctgatga tcttccacaa catggccatc ctggaggctt tgatgaccgt gcgtaagaag 1140 cgggccatct accccaatga cggcttcctg aagcagctgc gggagctcaa tgagaagttg 1200 atggaggaga gagaagagga ctatggccgg gaggggggat cagctgaggc tgaggagggc 1260 gagggcactg ggagcatgct cggggccaga gtgcacgccc tgacggtgga agaggaggac 1320 gacagcgcca gccacctgag tggctcctcc ctggggaagg ccacccaggc ctccaagccc 1380 ctcaccctca tagacgagga ggaggaggag aaactgtacg agcagtggaa gaaggggcag 1440 ggcctcctct cagacaaggt cccccaggat ggaggtggct ggcgctcagc ctcctctggc 1500 cagggtgggg aggagctcga ggacgaggac gtggagagga tcatccagga gtggcagagc 1560 cgaaacgaga ggtaccaagc agaagggtac cggaggtggg gaagggagga ggagaaggag 1620 gaggagagcg acgctggctc ctcggtgggg aggcggcggc gcaccctgag cgagagcagc 1680 gcctgggaga gcgtgagcag ccacgacatc tgggtcctga agcagcagct ggagctgaac 1740 cgcccggacc acggcaggag gcgccgcgca gactcgatgt cctcggagag cacctgggac 1800 gcatggaacg agaggctgct ggagattgag aaggaggctt cccggaggta ccacgccaag 1860 agcaagagag aggaggcggc agacaggagc tcagaagcag ggagcagggt gcgggaggat 1920 gatgaggaca gcgtgggctc tgaggccagt tccttctaca acttctgcag caggaacaag 1980 gacaagctca ctgccctgga aagatggaag atcaagagaa tccaatttgg atttcacaag 2040 aaagacttgg gagcgggaga cagcagcggt gagcccggtg cagaggaggc agtaggggag 2100 aagaacccct ccgacgtcag cctgacagcc taccaggcct ggaagctgaa acaccagaag 2160 aaggtgggca gtgagaacaa ggaggaggtg gtggagctca gcaaggggga ggactcggcc 2220 ttggctaaga agagacaacg gaggctggag ctgctggaga gaagccggca gacgctggag 2280 gagagccagt ctatggcaag ctgggaggcg gacagctcca cggccagcgg gagcattccc 2340 ctgtctgcgt tctggtctgc agacccctca gtcagcgctg atggggacac gacgtcagta 2400 ctgagcaccc agagccaccg ctcccacctg tctcaggctg caagcaacat agcggggtgt 2460 tcaacctcca accccaccac acccctgcct aacctgccag tggggcctgg agacaccatt 2520 tccattgcca gtatccagaa ctggattgcc aatgtagtca gtgagaccct tgctcagaag 2580 caaaatgaaa tgctgctgtt gtcccgctca ccgtctgttg caagcatgaa ggcagtacca 2640 gcggctagct gcctggggga tgaccaagtc tccatgctta gtggacacag cagctcctcc 2700 ttgggtggct gcctgttgcc tcagagccag gcaagaccca gctctgacat gcagtctgtg 2760 ctgtcctgca acaccacact gagctcaccc gcggaaagtt gcagaagcaa agtgaggggg 2820 accagcaagc ccatcttcag cctctttgct gacaatgtgg acctaaagga acttggccgg 2880 aaggagaagg agatgcagat ggagcttagg gagaagatgt ctgagtacca aatggaaaag 2940 ctggcctcag acaacaaacg cagctccctc ttcaagaaga agaaggtcaa ggaagatgag 3000 gatgatggtg tgggtgatgg ggatgaggac actgacagtg ccatagggag cttccgatat 3060 tcttcccgca gtaattccca gaaacctgaa acagacacat gctcctccct ggctgtctgt 3120 gatcactatg caagtggcag cagagttggc aaagagatgg atagcagtat taataagtgg 3180 ctcagtggcc tcaggacgga ggaaaaacct cctttccaaa gtgactggtc tggaagttcc 3240 agagggaagt acaccagatc gtccctgctc agggagacag agtctaaatc ctccagttac 3300 aagttttcca aatcccagtc agaggaacag gtacacctcc tcctaccacg aggcaaatgg 3360 caactctgta agaagcactt cacggttctc atcttcctcc accagggagg gcagagagat 3420 gcacaagttc tccaggtcca cgtacaacga gacctcaagt tcccgagagg agagcccaga 3480 gccctacttc ttccgccgga ccccagagtc ctcagaaagg gaagagtccc cagaaccaca 3540 gcgcccaaat tgggccaggt ccagggactg ggaagatgtg gaagagtcat ccaagtcaga 3600 cttctctgaa tttggagcca agaggaagtt cacccagagc tttatgaggt ctgaagaaga 3660 gggagagaaa gagaggacag aaaacagaga agaagggagg tttgcatctg gacggcggtc 3720 ccagtatcgg agaagcaatg acagggagga agaggaagaa atggacgatg aagccatcat 3780 tgctgcttgg agacgccggc aagaagaaac caggaccaag ctgcagaaaa ggagggagga 3840 ctgagctggg gaaaatctga gaacactgaa agaaaccact cacgttagca tagggctcag 3900 ggcacacgtt gccaccactc atcgcaggat gaggatacag agaggatctt ccagaggggc 3960 agagccaaaa tgagaggtac caagcataag ggcagcagag gtggagtagg gaggaggcaa 4020 ggagggggag aaccatcaat acgaatacga ggtccgaatg ctggaccaac tgataccatt 4080 ttctgttgct cagcgccctc taagctttgg tgtttcactt aatgtatttg acagtgttca 4140 tcacaggcta gagaggtgag cttggaaaag cactgtagtt tgtcagagac tccagtttac 4200 atccagaaag gccatgaaca taggacacgc ttctgtctgt agaggcttca tatgagaccc 4260 agaaagtcta tcctatggca agtctgacct ctcctggcaa tgctcagttc tgattttttt 4320 tttttaatgt tttgagtctc cattaaaaat ggtatgttgg 4360 5 1262 DNA Homo sapiens 5 gtcaagggtt tcaggtcgca ctggaaaatc attttgcaag cagatgtcat aggtctcctc 60 ttagactgga cggcacgcaa ggtcagcgtc acagatctga ccctaaaaat aggcctctgt 120 tgccagtcgg ggtggctggg cgtgcggctg ctacatgccc cacggaccag aacctcccga 180 cgcggccagg ccccggcaca cccagctgca gaaaggagag aaaatccctt ggctctaaaa 240 tgacatctgg agaagtgaag acaagcctca agaatgccta ctcatctgcc aagaggctgt 300 cgccgaagat ggaggaggaa ggggaggagg aggactactg cacccctgga gcctttgagc 360 tggagcggct cttctggaag ggcagtcccc agtacaccca cgtcaacgag gtctggccca 420 agctctacat tggcgatgag gcgacggcgc tggaccgcta taggctgcag aaggcggggt 480 tcacgcacgt gctgaacgcg gcccacggcc gctggaacgt ggacactggg cccgactact 540 accgcgacat ggacatccag taccacggcg tggaggccga cgacctgccc accttcgacc 600 tcagtgtctt cttctacccg gcggcagcct tcatcgacag agcgctaagc gacgaccaca 660 gtaagatcct ggttcactgc gtcatgggcc gcagccggtc agccaccctg gtcctggcct 720 acctgatgat ccacaaggac atgaccctgg tggacgccat ccagcaagtg gccaagaacc 780 gctgcgtcct cccgaaccgg ggctttttga agcagctccg ggagctggac aagcagctgg 840 tgcagcagag gcgacggtcc cagcgccagg acggtgagga ggaggatggc agggagctgt 900 aggcccgact cacagggcca gcagaggcac ttggggacag aggggagagg cagaacatag 960 ccctggccta ggactccaga gaagggatgg tgaaaccgaa gctcgactct tccaaaccat 1020 cttgttcaac ttccccatgt gtgctgggga cagggaggac ccagagctgc ccccgggcag 1080 agctgagcgc tcagcctctc agcaaaatgg gagggacggg ctccccggct ctgggtcaca 1140 gaggagcatg ccacgctgca ccaagtctcc tgctttggtt ttgttttttt ggtgagaagg 1200 aagagggaaa agattttaaa atgtggggct ttatttttgt aaatatcctt cgggctttgt 1260 tt 1262 6 1917 DNA Homo sapiens 6 acagaggcag aggggtgggc gggctggccc atggctgaga cctctctccc agagctgggg 60 ggagaggaca aagccacgcc ttgccccagc atcctggagc tggaggagct cctgcgggca 120 gggaagtctt cttgcagccg tgtggacgaa gtttggccca accttttcat aggagatgcg 180 gccacggcaa acaaccgctt tgagctgtgg aagctgggca tcacccacgt gctgaacgcc 240 gcccacaagg gcctctactg tcagggcggc cctgacttct acggcagcag tgtgagctac 300 ctgggggtgc cagcccacga cctccctgat tttgacatca gtgcctactt ctcctctgcg 360 gctgacttca tccaccgtgc cctcaacacg cctggggcca aggtcctggt gcactgtgtg 420 gtgggcgtga gccgctctgc cacgctggtc ctggcctacc tcatgctgca ccagcggctg 480 tccctgcgcc aggcggtgat caccgtgagg cagcaccgat gggtcttccc caaccgaggc 540 ttcctgcacc agctctgcag gctggaccac tggtccttac tccctgccat ggggctctgc 600 cactttgcca ccctggcact gatcctgctg gtgctgctgg aggctctggc ccaggcggac 660 acacagaaga tggtggaagc ccagcgtggg gtcggcccta gagcctgcta ctccatctgg 720 ctcctcctgg cgcctacacc ccctctcagc cactgtcttc agtctccaca gaaacagcat 780 caagtgtgcg gagacaggcg gctgaaagcc agcagcacga actgcccgtc agagaagtgc 840 acagcctggg ccagatactc ccacaggtgg gcccatattc tggtgccgct gaaaatccag 900 ctccgcaggg tccctgactc cttcagccag cagatgcctg aaacaagcta cctgacccgg 960 gtggggcctg acatccagtg ctggcctgag tcgtggggga tggactcact gcagaagcag 1020 gacctccgga ggcccaagat ccatggggca gtccaggcat ctccctacca gccgcccaca 1080 ttggcttcgc tgcagcgctt gctgtgggtc cgtcaggctg ccacactgaa ccatatcgat 1140 gaggtctggc ccagcctctt cctgggagat gcgtacgcag cccgggacaa gagcaagctg 1200 atccagctgg gaatcaccca cgttgtgaat gccgctgcag gcaagttcca ggtggacaca 1260 ggtgccaaat tctaccgtgg aatgtccctg gagtactatg gcatcgaggc ggacgacaac 1320 cccttcttcg acctcagtgt ctactttctg cctgttgctc gatacatccg agctgccctc 1380 agtgttcccc aagaggatgg ccatgggtgt ctcttcttcc caaaggggtg ggtggttcaa 1440 gggcaggtag ctgatgctaa gttggttctc cctacaggcc gcgtgctggt acactgtgcc 1500 atgggggtaa gccgctctgc cacacttgtc ctggccttcc tcatgatctg tgagaacatg 1560 acgctggtag aggccatcca gacggtgcag gcccaccgca atatctgccc taactcaggc 1620 ttcctccggc agctccaggt tctggacaac cgactggggc gggagacggg gcggttctga 1680 tctggcaggc agccaggatc cctgaccctt ggcccaaccc caccagcctg gccctgggaa 1740 cagcaggctc tgctgtttct agtgaccctg agatgtaaac agcaagtggg ggctgaggca 1800 gaggcaggga tagctgggtg gtgacctctt agcgggtgga tttccctgac ccaattcaga 1860 gattctttat gcaaaagtga gttcagtcca tctctataat aaaatattca tcgtcat 1917 7 636 DNA Homo sapiens 7 atgtgccctg gtaactggct ttgggcttct atgactttta tggcccgctt ctcccggagt 60 agctcaaggt ctcctgttcg aactcgaggg accctggagg agatgccaac cgttcaacat 120 cctttcctca atgtcttcga gttggagcgg ctcctctaca caggcaagac agcctgtaac 180 catgccgacg aggtctggcc aggcctctat ctcggagacc aggacatggc taacaaccgc 240 cgggagcttc gccgcctggg catcacgcac gtcctcaatg cctcacacag ccggtggcga 300 ggcacgcccg aggcctatga ggggctgggc atccgctacc tgggtgttga ggcccacgac 360 tcgccagcct ttgacatgag catccacttc cagacggctg ccgacttcat ccaccgggcg 420 ctgagccagc caggagggaa gatcctggtg cattgtgctg tgggcgtgag ccgatccgcc 480 accctggtac tggcctacct catgctgtac caccacctta ccctcgtgga ggccatcaag 540 aaagtcaaag accaccgagg catcatcccc aaccggggct tcctgaggca gctcctggcc 600 ctggaccgca ggctgcggca gggtctggaa gcatga 636 8 1326 DNA Homo sapiens 8 atgcaggggc agactgtagt tccaaaagat tcctacacta tatcccttat ccagaggctg 60 cggggccgtg aggccgcaag gagaacccat gagaaccttc ttcggctgtc tgccctagtg 120 agatccccac agacagctag catcgactgc cacacgtggt cagtttctag tggaaccaat 180 acttcgctgc aggcgtcggg cctgggccgt cagggcagct gtgaccggat cgcttcccgg 240 gcggcgagct gggggtgcac ccggaccgcc gcccccggga tcatgggcaa tggcatgacc 300 aaggtacttc ctggactcta cctcggaaac ttcattgatg ccaaagacct ggatcagctg 360 ggccgaaata agatcacaca catcatctct atccatgagt caccccagcc tctgctgcag 420 gatatcacct accttcgcat cccggtcgct gatacccctg aggtacccat caaaaagcac 480 ttcaaagaat gtatcaactt catccactgc tgccgcctta atggggggaa ctgccttgtg 540 cactgctttg caggcatctc tcgcagcacc acgattgtga cagcgtatgt gatgactgtg 600 acggggctag gctggcggga cgtgcttgaa gccatcaagg ccaccaggcc catcgccaac 660 cccaacccag gctttaggca gcagcttgaa gagtttggct gggccagttc ccagaagctt 720 cgccggcagc tggaggagcg cttcggcgag agccccttcc gcgacgagga ggagttgcgc 780 gcgctgctgc cgctgtgcaa gcgctgccgg cagggctccg cgacctcggc ctcctccgcc 840 gggccgcact cagcagcctc cgagggaacc gtgcagcgcc tggtgccgcg cacgccccgg 900 gaagcccacc ggccgctgcc gctgctggcg cgcgtcaagc agactttctc ttgcctcccc 960 cggtgtctgt cccgcaaggg cggcaagtga ggatgcagtc cagccgtggc tccccacttc 1020 cgactggctc ccttcggggg ctgtctgcgc cttccacgcc ccccaggacg ggcccagagg 1080 ctgggggagc cccgcggcgg cctgaaccct gcctcccgcg cccgccctgc tcgtccgcgt 1140 ctgcagtcag cgtccccaac ctgtgcgtct ctgtgtccgg gccggcctgc tgcagccacc 1200 tggtgcctta gtccttgggc tgggggaggg ggcccaccct taaaggcggc gggaggggag 1260 ggagggagag tggagggttt gacgggcctg gagggtatta aagagacaca gaagaagctg 1320 cctgtc 1326 9 1083 DNA Homo sapiens 9 atgatagagg atacaatgac tttgctgtct ctgctgggtc gcatcatgcg ctacttcttg 60 ctgagacccg agacgctttt cctgctgtgc atcagcttgg ctctatggag ttacttcttc 120 cacaccgacg aggtgaagac catcgtgaag tccagccggg acgccgtgaa gatggtgaag 180 agcaaggtag ccgagaccat gcagaacgat cgactcgggg ggcttgatgt gctcgaggcc 240 gagttttcca agacctggga gttcaagaac cacaacgtgg cggtgtactc catccagggc 300 cggagagacc acatggagga ccgcttcgaa gttctcacgg atctggccaa caagacgcac 360 ccgtccatct tcgggatctt cgacgggcac gggggagaga ctgcagctga atatgtaaaa 420 tctcgactcc cagaggctct taaacagcat cttcaggact acgagaaaga caaagaaaat 480 agtgtattat cttaccagac catccttgaa cagcagattt tgtcaattga ccgagaaatg 540 ctagaaaaat tgactgtatc ctatgatgaa gcaggcacaa cgtgtttgat tgctctgcta 600 tcagataaag acctcactgt ggccaacgtg ggtgactcgc gcggggtcct gtgtgacaaa 660 gatgggaacg ctattccttt gtctcatgat cacaagcctt accagttgaa ggaaagaaag 720 aggataaaga gagcaggtgg tttcatcagt ttcaatggct cctggagggt ccagggaatc 780 ctggccatgt ctcggtccct gggggattat ccgctgaaaa atctcaacgt ggtcatccca 840 gacccagaca tcctgacctt tgacctggac aagcttcagc ctgagttcat gatcttggca 900 tcagatggtc tctgggatgc tttcagcaat gaagaagcag ttcgattcat caaggagcgc 960 ttggatgaac ctcactttgg ggccaagagc atagttttac agtcatttta cagaggctgc 1020 cctgacaata taacagtcat ggtggtgaag ttcagaaata gcagcaaaac agaagagcag 1080 tga 1083 10 1725 DNA Homo sapiens 10 atgttgtcgg ctccgtgttg tgatgacagg agaatgtgtg tgtgtcccgg gcccagacga 60 attggaatcc cagtcagaag ttccagcctg ccactgttct ctgatgccat gccagcacca 120 actcaactgt tttttcctct catccgtaac tgtgaactga gcaggatcta tggcactgca 180 tgttactgcc accacaaaca tctctgttgt tcctcatcgt acattcctca gagtcgactg 240 agatacacac ctcatccagc atatgctacc ttttgcaggc caaaggagaa ctggtggcag 300 tacacccaag gaaggagata tgcttccaca ccacagaaat tttacctcac acctccacaa 360 gtcaatagca tccttaaagc taatgaatac agtttcaaag tgccagaatt tgacggcaaa 420 atgtcagttc tatccttgga tttgacagca atcaagctgc ctgcaaatgc acccattgag 480 gaccggagaa gtgcagcaac ctgcttgcag accagaggga tgcttttggg ggtttttgat 540 ggccatgcag gttgtgcttg gtcccaggca gtcagtgaaa gactctttta ttatattgct 600 ggctctttgg taccccatga gactttgcta gagattgaaa atgcagtgga gagcggccgg 660 gcactgctac ccattctcca gtggcacaag caccccaatg attactttag taaggaggca 720 tccaaattgt actttaacag cttgaggact tactggcaag agcttataga cctcaacact 780 ggtgagtcga ctgatattga tgttaaggag gctctaatta atgccttcaa gaggcttgat 840 aatgacatct ccttggaggc gcaagttggt gatcctaatt cttttctcaa ctacctggtg 900 cttcgagtgg cattttctgg agccactgct tgtgtggccc atgtggatgg tgttgacctt 960 catgtggcca atactggcga tagcagagcc atgctgggtg tgcaggaaga ggacggctca 1020 tggtcagcag tcacgctgtc taatgaccac aatgctcaaa atgaaagaga actagaacgg 1080 ctgaaattgg aacatccaaa gagtgaggcc aagagtgtcg tgaaacagga tcggctgctt 1140 ggcttgctga tgccatttag ggcatttgga gatgtaaagt tcaaatggag cattgacctt 1200 caaaagagag tgatagaatc tggcccagac cagttgaatg acaatgaata taccaagttt 1260 attcctccta attatcacac acctccttat ctcactgctg agccagaggt aacttaccac 1320 cgattaaggc cacaggataa gtttctggtg ttggctactg atgggttgtg ggagactatg 1380 cataggcagg atgtggttag gattgtgggt gagtacctaa ctggcatgca tcaccaacag 1440 ccaatagctg ttggtggcta caaggtgact ctgggacaga tgcatggcct tttaacagaa 1500 aggagaacca aaatgtcctc ggtatttgag gatcagaacg cagcaaccca tctcattcgc 1560 cacgctgtgg gcaacaacga gtttgggact gttgatcatg agcgcctctc taaaatgctt 1620 agtcttcctg aagagcttgc tcgaatgtac agagatgaca ttacaatcat tgtagttcag 1680 ttcaattctc atgttgtagg ggcgtatcaa aaccaagaaa agtga 1725 11 909 DNA Homo sapiens 11 atgaggctcc caatcctgtt cgctgccctg ctctggttcc ggggttttct ggcagaggag 60 gaagcatgcc tctccctgga agggagtcca ggcagggaga gtgcaggccc acccgtgaac 120 gtgaacatca ccagccaggg gagacctact agcctctttc tgagctgggc agccccgggg 180 ccaggcaggt tcacccatgc cctccgcctc acatgtctga gccccctcag ctctcctgaa 240 gggcagcagc tccaggccca caccaatgca tccagcttta agttccaaga tctggtgtca 300 gggggtcgct accagctgga agtgactgcc ctgcgaccct gtgggcagaa tgtcaccatc 360 accctcactg ctcgcactgc cccgtcaact gtccatggac tgcagctcca ctctgggagc 420 ccatccagcc tggaggcctc atggggtgat gcccccggga agcaggatgg ctactgcctt 480 ctcctctacc acctagaatc ccagacattg gcacataata tctccatgcc cctgggcacc 540 ctgtcctaca attttggcaa cctcttgcca ggtattgagt atattttgga agttaacacc 600 tgggctggca acctccaagc aacaaccagc ctccatcagt ggacagcccc tgtgtctcca 660 gatcacctgg ttctgcatac cctgggcacc agtgccttgc aagcctcctg gaacggctcc 720 aagggggctg cctggctcca cttggtgctc acagacctac ctggtggcac caatctgact 780 gcagtattca gacggggagt ctcccatcac acctcccttc acctgtctca gggccccccc 840 tatgagctga cgctcagtgc tgctgccagg ccccatcggg cagtggggcc caatgccaca 900 gagtggacc 909 12 354 DNA Homo sapiens modified_base (1)..(354) “n” represents a, t, c , g, other or unknown 12 tctgaaggag ttggctggac tggttgtttc attgtcatag atgccatgtt ggaaagaatc 60 aagcatgaaa aaactgtagg taattatgcc tatgcaactt taatgagaac ccagaggaat 120 tacatggttc aagcaggaga ccagtgtatc tctgtccatg atgcactgtt agaggcagtt 180 acttgtgtaa ataccaaagt tccagctaga aacttgtatg cctatattan gaaactgaca 240 caaatagaga ggggacagaa tgtcatagga gtggtgctca aatttaagca tctaatcagc 300 tcaaaagctc acatctcagg tttcctcagt gccaatcttc catgcaataa tttc 354 13 1049 PRT Homo sapiens 13 Met Ala Leu Val Thr Leu Gln Arg Ser Pro Thr Pro Ser Ala Ala Ser 1 5 10 15 Ser Ser Ala Ser Asn Ser Glu Leu Glu Ala Gly Ser Glu Glu Asp Arg 20 25 30 Lys Leu Asn Leu Ser Leu Ser Glu Ser Phe Phe Met Val Lys Gly Ala 35 40 45 Ala Leu Phe Leu Gln Gln Gly Ser Ser Pro Gln Gly Gln Arg Ser Leu 50 55 60 Gln His Pro His Lys His Ala Gly Asp Leu Pro Gln His Leu Gln Val 65 70 75 80 Met Ile Asn Leu Leu Arg Cys Glu Asp Arg Ile Lys Leu Ala Val Arg 85 90 95 Leu Glu Ser Ala Trp Ala Asp Arg Val Arg Tyr Met Val Val Val Tyr 100 105 110 Ser Ser Gly Arg Gln Asp Thr Glu Glu Asn Ile Leu Leu Gly Val Asp 115 120 125 Phe Ser Ser Lys Glu Ser Lys Ser Cys Thr Ile Gly Met Val Leu Arg 130 135 140 Leu Trp Ser Asp Thr Lys Ile His Leu Asp Gly Asp Gly Gly Phe Ser 145 150 155 160 Val Ser Thr Ala Gly Arg Met His Ile Phe Lys Pro Val Ser Val Gln 165 170 175 Ala Met Trp Ser Ala Leu Gln Val Leu His Lys Ala Cys Glu Val Ala 180 185 190 Arg Arg His Asn Tyr Phe Pro Gly Gly Val Ala Leu Ile Trp Ala Thr 195 200 205 Tyr Tyr Glu Ser Cys Ile Ser Ser Glu Gln Ser Cys Ile Asn Glu Trp 210 215 220 Asn Ala Met Gln Asp Leu Glu Ser Thr Arg Pro Asp Ser Pro Ala Leu 225 230 235 240 Phe Val Asp Lys Pro Thr Glu Gly Glu Arg Thr Glu Arg Leu Ile Lys 245 250 255 Ala Lys Leu Arg Ser Ile Met Met Ser Gln Asp Leu Glu Asn Val Thr 260 265 270 Ser Lys Glu Ile Arg Asn Glu Leu Glu Lys Gln Met Asn Cys Asn Leu 275 280 285 Lys Glu Leu Lys Glu Phe Ile Asp Asn Glu Met Leu Leu Ile Leu Gly 290 295 300 Gln Met Asp Lys Pro Ser Leu Ile Phe Asp His Leu Tyr Leu Gly Ser 305 310 315 320 Glu Trp Asn Ala Ser Asn Leu Glu Glu Leu Gln Gly Ser Gly Val Asp 325 330 335 Tyr Ile Leu Asn Val Thr Arg Glu Ile Asp Asn Phe Phe Pro Gly Leu 340 345 350 Phe Ala Tyr His Asn Ile Arg Val Tyr Asp Glu Glu Thr Thr Asp Leu 355 360 365 Leu Ala His Trp Asn Glu Ala Tyr His Phe Ile Asn Lys Ala Lys Arg 370 375 380 Asn His Ser Lys Cys Leu Val His Cys Lys Met Gly Val Ser Arg Ser 385 390 395 400 Ala Ser Thr Val Ile Ala Tyr Ala Met Lys Glu Phe Gly Trp Pro Leu 405 410 415 Glu Lys Ala Tyr Asn Tyr Val Lys Gln Lys Arg Ser Ile Thr Arg Pro 420 425 430 Asn Ala Gly Phe Met Arg Gln Leu Ser Glu Tyr Glu Gly Ile Leu Asp 435 440 445 Ala Ser Lys Gln Arg His Asn Lys Leu Trp Arg Gln Gln Thr Asp Ser 450 455 460 Ser Leu Gln Gln Pro Val Asp Asp Pro Ala Gly Pro Gly Asp Phe Leu 465 470 475 480 Pro Glu Thr Pro Asp Gly Thr Pro Glu Ser Gln Leu Pro Phe Leu Asp 485 490 495 Asp Ala Ala Gln Pro Gly Leu Gly Pro Pro Leu Pro Cys Cys Phe Arg 500 505 510 Arg Leu Ser Asp Pro Leu Leu Pro Ser Pro Glu Asp Glu Thr Gly Ser 515 520 525 Leu Val His Leu Glu Asp Pro Glu Arg Glu Ala Leu Leu Glu Glu Ala 530 535 540 Ala Pro Pro Ala Glu Val His Arg Pro Ala Arg Gln Pro Gln Gln Gly 545 550 555 560 Ser Gly Leu Cys Glu Lys Asp Val Lys Lys Lys Leu Glu Phe Gly Ser 565 570 575 Pro Lys Gly Arg Ser Gly Ser Leu Leu Gln Val Glu Glu Thr Glu Arg 580 585 590 Glu Glu Gly Leu Gly Ala Gly Arg Trp Gly Gln Leu Pro Thr Gln Leu 595 600 605 Asp Gln Asn Leu Leu Asn Ser Glu Asn Leu Asn Asn Asn Ser Lys Arg 610 615 620 Ser Cys Pro Asn Gly Met Glu Asp Asp Ala Ile Phe Gly Ile Leu Asn 625 630 635 640 Lys Val Lys Pro Ser Tyr Lys Ser Cys Ala Asp Cys Met Tyr Pro Thr 645 650 655 Ala Ser Gly Ala Pro Glu Ala Ser Arg Glu Arg Cys Glu Asp Pro Asn 660 665 670 Ala Pro Ala Ile Cys Thr Gln Pro Ala Phe Leu Pro His Ile Thr Ser 675 680 685 Ser Pro Val Ala His Leu Ala Ser Arg Ser Arg Val Pro Glu Lys Pro 690 695 700 Ala Ser Gly Pro Thr Glu Pro Pro Pro Phe Leu Pro Pro Ala Gly Ser 705 710 715 720 Arg Arg Ala Asp Thr Ser Gly Pro Gly Ala Gly Ala Ala Leu Glu Pro 725 730 735 Pro Ala Ser Leu Leu Glu Pro Ser Arg Glu Thr Pro Lys Val Leu Pro 740 745 750 Lys Ser Leu Leu Leu Lys Asn Ser His Cys Asp Lys Asn Pro Pro Ser 755 760 765 Thr Glu Val Val Ile Lys Glu Glu Ser Ser Pro Lys Lys Asp Met Lys 770 775 780 Pro Ala Lys Asp Leu Arg Leu Leu Phe Ser Asn Glu Ser Glu Lys Pro 785 790 795 800 Thr Thr Asn Ser Tyr Leu Met Gln His Gln Glu Ser Ile Ile Gln Leu 805 810 815 Gln Lys Ala Gly Leu Val Arg Lys His Thr Lys Glu Leu Glu Arg Leu 820 825 830 Lys Ser Val Pro Ala Asp Pro Ala Pro Pro Ser Arg Asp Gly Pro Ala 835 840 845 Ser Arg Leu Glu Ala Ser Ile Pro Glu Glu Ser Gln Asp Pro Ala Ala 850 855 860 Leu His Glu Leu Gly Pro Leu Val Met Pro Ser Gln Ala Gly Ser Asp 865 870 875 880 Glu Lys Ser Glu Ala Ala Pro Ala Ser Leu Glu Gly Gly Ser Leu Lys 885 890 895 Ser Pro Pro Pro Phe Phe Tyr Arg Leu Asp His Thr Ser Ser Phe Ser 900 905 910 Lys Asp Phe Leu Lys Thr Ile Cys Tyr Thr Pro Thr Ser Ser Ser Met 915 920 925 Ser Ser Asn Leu Thr Arg Ser Ser Ser Ser Asp Ser Ile His Ser Val 930 935 940 Arg Gly Lys Pro Gly Leu Val Lys Gln Arg Thr Gln Glu Ile Glu Thr 945 950 955 960 Arg Leu Arg Leu Ala Gly Leu Thr Val Ser Ser Pro Leu Lys Arg Ser 965 970 975 His Ser Leu Ala Lys Leu Gly Ser Leu Thr Phe Ser Thr Glu Asp Leu 980 985 990 Ser Ser Glu Ala Asp Pro Ser Thr Val Ala Asp Ser Gln Asp Thr Thr 995 1000 1005 Leu Ser Glu Ser Ser Phe Leu His Glu Pro Gln Gly Thr Pro Arg Asp 1010 1015 1020 Pro Ala Ala Thr Ser Lys Pro Ser Gly Lys Pro Ala Pro Glu Asn Leu 1025 1030 1035 1040 Lys Ser Pro Ser Trp Met Ser Lys Ser 1045 14 665 PRT Homo sapiens 14 Met Ala His Glu Met Ile Gly Thr Gln Ile Val Thr Glu Arg Leu Val 1 5 10 15 Ala Leu Leu Glu Ser Gly Thr Glu Lys Val Leu Leu Ile Asp Ser Arg 20 25 30 Pro Phe Val Glu Tyr Asn Thr Ser His Ile Leu Glu Ala Ile Asn Ile 35 40 45 Asn Cys Ser Lys Leu Met Lys Arg Arg Leu Gln Gln Asp Lys Val Leu 50 55 60 Ile Thr Glu Leu Ile Gln His Ser Ala Lys His Lys Val Asp Ile Asp 65 70 75 80 Cys Ser Gln Lys Val Val Val Tyr Asp Gln Ser Ser Gln Asp Val Ala 85 90 95 Ser Leu Ser Ser Asp Cys Phe Leu Thr Val Leu Leu Gly Lys Leu Glu 100 105 110 Lys Ser Phe Asn Ser Val His Leu Leu Ala Gly Gly Phe Ala Glu Phe 115 120 125 Ser Arg Cys Phe Pro Gly Leu Cys Glu Gly Lys Ser Thr Leu Val Pro 130 135 140 Thr Cys Ile Ser Gln Pro Cys Leu Pro Val Ala Asn Ile Gly Pro Thr 145 150 155 160 Arg Ile Leu Pro Asn Leu Tyr Leu Gly Cys Gln Arg Asp Val Leu Asn 165 170 175 Lys Glu Leu Met Gln Gln Asn Gly Ile Gly Tyr Val Leu Asn Ala Ser 180 185 190 Asn Thr Cys Pro Lys Pro Asp Phe Ile Pro Glu Ser His Phe Leu Arg 195 200 205 Val Pro Val Asn Asp Ser Phe Cys Glu Lys Ile Leu Pro Trp Leu Asp 210 215 220 Lys Ser Val Asp Phe Ile Glu Lys Ala Lys Ala Ser Asn Gly Cys Val 225 230 235 240 Leu Val His Cys Leu Ala Gly Ile Ser Arg Ser Ala Thr Ile Ala Ile 245 250 255 Ala Tyr Ile Met Lys Arg Met Asp Met Ser Leu Asp Glu Ala Tyr Arg 260 265 270 Phe Val Lys Glu Lys Arg Pro Thr Ile Ser Pro Asn Phe Asn Phe Leu 275 280 285 Gly Gln Leu Leu Asp Tyr Glu Lys Lys Ile Lys Asn Gln Thr Gly Ala 290 295 300 Ser Gly Pro Lys Ser Lys Leu Lys Leu Leu His Leu Glu Lys Pro Asn 305 310 315 320 Glu Pro Val Pro Ala Val Ser Glu Gly Gly Gln Lys Ser Glu Thr Pro 325 330 335 Leu Ser Pro Pro Cys Ala Asp Ser Ala Thr Ser Glu Ala Ala Gly Gln 340 345 350 Arg Pro Val His Pro Ala Ser Val Pro Ser Val Pro Ser Val Gln Pro 355 360 365 Ser Leu Leu Glu Asp Ser Pro Leu Val Gln Ala Leu Ser Gly Leu His 370 375 380 Leu Ser Ala Asp Arg Leu Glu Asp Ser Asn Lys Leu Lys Arg Ser Phe 385 390 395 400 Ser Leu Asp Ile Lys Ser Val Ser Tyr Ser Ala Ser Met Ala Ala Ser 405 410 415 Leu His Gly Phe Ser Ser Ser Glu Asp Ala Leu Glu Tyr Tyr Lys Pro 420 425 430 Ser Thr Thr Leu Asp Gly Thr Asn Lys Leu Cys Gln Phe Ser Pro Val 435 440 445 Gln Glu Leu Ser Glu Gln Thr Pro Glu Thr Ser Pro Asp Lys Glu Glu 450 455 460 Ala Ser Ile Pro Lys Lys Leu Gln Thr Ala Arg Pro Ser Asp Ser Gln 465 470 475 480 Ser Lys Arg Leu His Ser Val Arg Thr Ser Ser Ser Gly Thr Ala Gln 485 490 495 Arg Ser Leu Leu Ser Pro Leu His Arg Ser Gly Ser Val Glu Asp Asn 500 505 510 Tyr His Thr Ser Phe Leu Phe Gly Leu Ser Thr Ser Gln Gln His Leu 515 520 525 Thr Lys Ser Ala Gly Leu Gly Leu Lys Gly Trp His Ser Asp Ile Leu 530 535 540 Ala Pro Gln Thr Ser Thr Pro Ser Leu Thr Ser Ser Trp Tyr Phe Ala 545 550 555 560 Thr Glu Ser Ser His Phe Tyr Ser Ala Ser Ala Ile Tyr Gly Gly Ser 565 570 575 Ala Ser Tyr Ser Ala Tyr Ser Cys Ser Gln Leu Pro Thr Cys Gly Asp 580 585 590 Gln Val Tyr Ser Val Arg Arg Arg Gln Lys Pro Ser Asp Arg Ala Asp 595 600 605 Ser Arg Arg Ser Trp His Glu Glu Ser Pro Phe Glu Lys Gln Phe Lys 610 615 620 Arg Arg Ser Cys Gln Met Glu Phe Gly Glu Ser Ile Met Ser Glu Asn 625 630 635 640 Arg Ser Arg Glu Glu Leu Gly Lys Val Gly Ser Gln Ser Ser Phe Ser 645 650 655 Gly Ser Met Glu Ile Ile Glu Val Ser 660 665 15 498 PRT Homo sapiens 15 Met Ala Leu Val Thr Val Gln Arg Ser Pro Thr Pro Ser Thr Thr Ser 1 5 10 15 Ser Pro Cys Ala Ser Glu Ala Asp Ser Gly Glu Glu Glu Cys Arg Ser 20 25 30 Gln Pro Arg Ser Ile Ser Glu Ser Phe Leu Thr Val Lys Gly Ala Ala 35 40 45 Leu Phe Leu Pro Arg Gly Asn Gly Ser Ser Thr Pro Arg Ile Ser His 50 55 60 Arg Arg Asn Lys His Ala Gly Asp Leu Gln Gln His Leu Gln Ala Met 65 70 75 80 Phe Ile Leu Leu Arg Pro Glu Asp Asn Ile Arg Leu Ala Val Arg Leu 85 90 95 Glu Ser Thr Tyr Gln Asn Arg Thr Arg Tyr Met Val Val Val Ser Thr 100 105 110 Asn Gly Arg Gln Asp Thr Glu Glu Ser Ile Val Leu Gly Met Asp Phe 115 120 125 Ser Ser Asn Asp Ser Ser Thr Cys Thr Met Gly Leu Val Leu Pro Leu 130 135 140 Trp Ser Asp Thr Leu Ile His Leu Asp Gly Asp Gly Gly Phe Ser Val 145 150 155 160 Ser Thr Asp Asn Arg Val His Ile Phe Lys Pro Val Ser Val Gln Ala 165 170 175 Met Trp Ser Ala Leu Gln Ser Leu His Lys Ala Cys Glu Val Ala Arg 180 185 190 Ala His Asn Tyr Tyr Pro Gly Ser Leu Phe Leu Thr Trp Val Ser Tyr 195 200 205 Tyr Glu Ser His Ile Asn Ser Asp Gln Ser Ser Val Asn Glu Trp Asn 210 215 220 Ala Met Gln Asp Val Gln Ser His Arg Pro Asp Ser Pro Ala Leu Phe 225 230 235 240 Thr Asp Ile Pro Thr Glu Arg Glu Arg Thr Glu Arg Leu Ile Lys Thr 245 250 255 Lys Leu Arg Glu Ile Met Met Gln Lys Asp Leu Glu Asn Ile Thr Ser 260 265 270 Lys Glu Ile Arg Thr Glu Leu Glu Met Gln Met Val Cys Asn Leu Arg 275 280 285 Glu Phe Lys Glu Phe Ile Asp Asn Glu Met Ile Val Ile Leu Gly Gln 290 295 300 Met Asp Ser Pro Thr Gln Ile Phe Glu His Val Phe Leu Gly Ser Glu 305 310 315 320 Trp Asn Ala Ser Asn Leu Glu Asp Leu Gln Asn Arg Gly Val Arg Tyr 325 330 335 Ile Leu Asn Val Thr Arg Glu Ile Asp Asn Phe Phe Pro Gly Val Phe 340 345 350 Glu Tyr His Asn Ile Arg Val Tyr Asp Glu Glu Ala Thr Asp Leu Leu 355 360 365 Ala Tyr Trp Asn Asp Thr Tyr Lys Phe Ile Ser Lys Ala Lys Lys His 370 375 380 Gly Ser Lys Cys Leu Val His Cys Lys Met Gly Val Ser Arg Ser Ala 385 390 395 400 Ser Thr Val Ile Ala Tyr Ala Met Lys Glu Tyr Asp Arg Ala Tyr Asp 405 410 415 Tyr Val Lys Glu Arg Arg Thr Val Thr Lys Pro Asn Pro Ser Phe Met 420 425 430 Arg Gln Leu Glu Glu Tyr Gln Gly Ile Leu Leu Ala Ser Phe Leu Gly 435 440 445 Leu Ile His Gly Gly Arg Asp Lys Pro Trp Gly Glu Lys Ser Thr Glu 450 455 460 Phe Glu Ser Val Asp Leu Val Ser Ile Pro Gly Ser Pro Ser Cys Cys 465 470 475 480 Asn Pro Glu Lys Leu Leu His Ile Ser His Pro Tyr Leu Thr Pro Ser 485 490 495 Ile Lys 16 1133 PRT Homo sapiens 16 Met Met Ala Gly Thr Ser Cys Trp Tyr Pro Ser Cys Pro Leu Ile Gly 1 5 10 15 Ser Arg Met Thr Pro Glu Pro Lys Ala Leu Pro Pro Val Ala Leu Val 20 25 30 Arg Asp Thr Glu Gly Gln Leu Cys Leu Pro Gln Gln Arg Gln Arg Gly 35 40 45 Trp Gln Val Val Met Ala Thr Arg Lys Asp Thr Glu Glu Glu Gln Val 50 55 60 Val Pro Ser Glu Glu Asp Glu Ala Asn Val Arg Ala Val Gln Ala His 65 70 75 80 Tyr Leu Arg Ser Pro Ser Pro Ser Gln Tyr Ser Met Val Ser Asp Ala 85 90 95 Glu Thr Glu Ser Ile Phe Met Glu Pro Ile His Leu Ser Ser Ala Ile 100 105 110 Ala Ala Lys Gln Ile Ile Asn Glu Glu Leu Lys Pro Pro Gly Val Arg 115 120 125 Ala Asp Ala Glu Cys Pro Gly Met Leu Glu Ser Ala Glu Gln Leu Leu 130 135 140 Val Glu Asp Leu Tyr Asn Arg Val Arg Glu Lys Met Asp Asp Thr Ser 145 150 155 160 Leu Tyr Asn Thr Pro Cys Val Leu Asp Leu Gln Arg Ala Leu Val Gln 165 170 175 Asp Arg Gln Glu Ala Pro Trp Asn Glu Val Asp Glu Val Trp Pro Asn 180 185 190 Val Phe Ile Ala Glu Lys Ser Val Ala Val Asn Lys Gly Arg Leu Lys 195 200 205 Arg Leu Gly Ile Thr His Ile Leu Asn Ala Ala His Gly Thr Gly Val 210 215 220 Tyr Thr Gly Pro Glu Phe Tyr Thr Gly Leu Glu Ile Gln Tyr Leu Gly 225 230 235 240 Val Glu Val Asp Asp Phe Pro Glu Val Asp Ile Ser Gln His Phe Arg 245 250 255 Lys Ala Tyr Cys His Tyr Ile Ile Phe Ser Cys Val Phe Ile Ser Gly 260 265 270 Lys Val Leu Val Ser Ser Glu Met Gly Ile Ser Arg Ser Ala Val Leu 275 280 285 Val Val Ala Tyr Leu Met Ile Phe His Asn Met Ala Ile Leu Glu Ala 290 295 300 Leu Met Thr Val Arg Lys Lys Arg Ala Ile Tyr Pro Asn Asp Gly Phe 305 310 315 320 Leu Lys Gln Leu Arg Glu Leu Asn Glu Lys Leu Met Glu Glu Arg Glu 325 330 335 Glu Asp Tyr Gly Arg Glu Gly Gly Ser Ala Glu Ala Glu Glu Gly Glu 340 345 350 Gly Thr Gly Ser Met Leu Gly Ala Arg Val His Ala Leu Thr Val Glu 355 360 365 Glu Glu Asp Asp Ser Ala Ser His Leu Ser Gly Ser Ser Leu Gly Lys 370 375 380 Ala Thr Gln Ala Ser Lys Pro Leu Thr Leu Ile Asp Glu Glu Glu Glu 385 390 395 400 Glu Lys Leu Tyr Glu Gln Trp Lys Lys Gly Gln Gly Leu Leu Ser Asp 405 410 415 Lys Val Pro Gln Asp Gly Gly Gly Trp Arg Ser Ala Ser Ser Gly Gln 420 425 430 Gly Gly Glu Glu Leu Glu Asp Glu Asp Val Glu Arg Ile Ile Gln Glu 435 440 445 Trp Gln Ser Arg Asn Glu Arg Tyr Gln Ala Glu Gly Tyr Arg Arg Trp 450 455 460 Gly Arg Glu Glu Glu Lys Glu Glu Glu Ser Asp Ala Gly Ser Ser Val 465 470 475 480 Gly Arg Arg Arg Arg Thr Leu Ser Glu Ser Ser Ala Trp Glu Ser Val 485 490 495 Ser Ser His Asp Ile Trp Val Leu Lys Gln Gln Leu Glu Leu Asn Arg 500 505 510 Pro Asp His Gly Arg Arg Arg Arg Ala Asp Ser Met Ser Ser Glu Ser 515 520 525 Thr Trp Asp Ala Trp Asn Glu Arg Leu Leu Glu Ile Glu Lys Glu Ala 530 535 540 Ser Arg Arg Tyr His Ala Lys Ser Lys Arg Glu Glu Ala Ala Asp Arg 545 550 555 560 Ser Ser Glu Ala Gly Ser Arg Val Arg Glu Asp Asp Glu Asp Ser Val 565 570 575 Gly Ser Glu Ala Ser Ser Phe Tyr Asn Phe Cys Ser Arg Asn Lys Asp 580 585 590 Lys Leu Thr Ala Leu Glu Arg Trp Lys Ile Lys Arg Ile Gln Phe Gly 595 600 605 Phe His Lys Lys Asp Leu Gly Ala Gly Asp Ser Ser Gly Glu Pro Gly 610 615 620 Ala Glu Glu Ala Val Gly Glu Lys Asn Pro Ser Asp Val Ser Leu Thr 625 630 635 640 Ala Tyr Gln Ala Trp Lys Leu Lys His Gln Lys Lys Val Gly Ser Glu 645 650 655 Asn Lys Glu Glu Val Val Glu Leu Ser Lys Gly Glu Asp Ser Ala Leu 660 665 670 Ala Lys Lys Arg Gln Arg Arg Leu Glu Leu Leu Glu Arg Ser Arg Gln 675 680 685 Thr Leu Glu Glu Ser Gln Ser Met Ala Ser Trp Glu Ala Asp Ser Ser 690 695 700 Thr Ala Ser Gly Ser Ile Pro Leu Ser Ala Phe Trp Ser Ala Asp Pro 705 710 715 720 Ser Val Ser Ala Asp Gly Asp Thr Thr Ser Val Leu Ser Thr Gln Ser 725 730 735 His Arg Ser His Leu Ser Gln Ala Ala Ser Asn Ile Ala Gly Cys Ser 740 745 750 Thr Ser Asn Pro Thr Thr Pro Leu Pro Asn Leu Pro Val Gly Pro Gly 755 760 765 Asp Thr Ile Ser Ile Ala Ser Ile Gln Asn Trp Ile Ala Asn Val Val 770 775 780 Ser Glu Thr Leu Ala Gln Lys Gln Asn Glu Met Leu Leu Leu Ser Arg 785 790 795 800 Ser Pro Ser Val Ala Ser Met Lys Ala Val Pro Ala Ala Ser Cys Leu 805 810 815 Gly Asp Asp Gln Val Ser Met Leu Ser Gly His Ser Ser Ser Ser Leu 820 825 830 Gly Gly Cys Leu Leu Pro Gln Ser Gln Ala Arg Pro Ser Ser Asp Met 835 840 845 Gln Ser Val Leu Ser Cys Asn Thr Thr Leu Ser Ser Pro Ala Glu Ser 850 855 860 Cys Arg Ser Lys Val Arg Gly Thr Ser Lys Pro Ile Phe Ser Leu Phe 865 870 875 880 Ala Asp Asn Val Asp Leu Lys Glu Leu Gly Arg Lys Glu Lys Glu Met 885 890 895 Gln Met Glu Leu Arg Glu Lys Met Ser Glu Tyr Gln Met Glu Lys Leu 900 905 910 Ala Ser Asp Asn Lys Arg Ser Ser Leu Phe Lys Lys Lys Lys Val Lys 915 920 925 Glu Asp Glu Asp Asp Gly Val Gly Asp Gly Asp Glu Asp Thr Asp Ser 930 935 940 Ala Ile Gly Ser Phe Arg Tyr Ser Ser Arg Ser Asn Ser Gln Lys Pro 945 950 955 960 Glu Thr Asp Thr Cys Ser Ser Leu Ala Val Cys Asp His Tyr Ala Ser 965 970 975 Gly Ser Arg Val Gly Lys Glu Met Asp Ser Ser Ile Asn Lys Trp Leu 980 985 990 Ser Gly Leu Arg Thr Glu Glu Lys Pro Pro Phe Gln Ser Asp Trp Ser 995 1000 1005 Gly Ser Ser Arg Gly Lys Tyr Thr Arg Ser Ser Leu Leu Arg Glu Thr 1010 1015 1020 Glu Ser Lys Ser Ser Ser Tyr Lys Phe Ser Lys Ser Gln Ser Glu Glu 1025 1030 1035 1040 Gln Val His Leu Leu Leu Pro Arg Gly Lys Trp Gln Leu Cys Lys Lys 1045 1050 1055 His Phe Thr Val Leu Ile Phe Leu His Gln Gly Gly Gln Arg Asp Ala 1060 1065 1070 Gln Val Leu Gln Val His Val Gln Arg Asp Leu Lys Phe Pro Arg Gly 1075 1080 1085 Glu Pro Arg Ala Leu Leu Leu Pro Pro Asp Pro Arg Val Leu Arg Lys 1090 1095 1100 Gly Arg Val Pro Arg Thr Thr Ala Pro Lys Leu Gly Gln Val Gln Gly 1105 1110 1115 1120 Leu Gly Arg Cys Gly Arg Val Ile Gln Val Arg Leu Leu 1125 1130 17 220 PRT Homo sapiens 17 Met Thr Ser Gly Glu Val Lys Thr Ser Leu Lys Asn Ala Tyr Ser Ser 1 5 10 15 Ala Lys Arg Leu Ser Pro Lys Met Glu Glu Glu Gly Glu Glu Glu Asp 20 25 30 Tyr Cys Thr Pro Gly Ala Phe Glu Leu Glu Arg Leu Phe Trp Lys Gly 35 40 45 Ser Pro Gln Tyr Thr His Val Asn Glu Val Trp Pro Lys Leu Tyr Ile 50 55 60 Gly Asp Glu Ala Thr Ala Leu Asp Arg Tyr Arg Leu Gln Lys Ala Gly 65 70 75 80 Phe Thr His Val Leu Asn Ala Ala His Gly Arg Trp Asn Val Asp Thr 85 90 95 Gly Pro Asp Tyr Tyr Arg Asp Met Asp Ile Gln Tyr His Gly Val Glu 100 105 110 Ala Asp Asp Leu Pro Thr Phe Asp Leu Ser Val Phe Phe Tyr Pro Ala 115 120 125 Ala Ala Phe Ile Asp Arg Ala Leu Ser Asp Asp His Ser Lys Ile Leu 130 135 140 Val His Cys Val Met Gly Arg Ser Arg Ser Ala Thr Leu Val Leu Ala 145 150 155 160 Tyr Leu Met Ile His Lys Asp Met Thr Leu Val Asp Ala Ile Gln Gln 165 170 175 Val Ala Lys Asn Arg Cys Val Leu Pro Asn Arg Gly Phe Leu Lys Gln 180 185 190 Leu Arg Glu Leu Asp Lys Gln Leu Val Gln Gln Arg Arg Arg Ser Gln 195 200 205 Arg Gln Asp Gly Glu Glu Glu Asp Gly Arg Glu Leu 210 215 220 18 549 PRT Homo sapiens 18 Met Ala Glu Thr Ser Leu Pro Glu Leu Gly Gly Glu Asp Lys Ala Thr 1 5 10 15 Pro Cys Pro Ser Ile Leu Glu Leu Glu Glu Leu Leu Arg Ala Gly Lys 20 25 30 Ser Ser Cys Ser Arg Val Asp Glu Val Trp Pro Asn Leu Phe Ile Gly 35 40 45 Asp Ala Ala Thr Ala Asn Asn Arg Phe Glu Leu Trp Lys Leu Gly Ile 50 55 60 Thr His Val Leu Asn Ala Ala His Lys Gly Leu Tyr Cys Gln Gly Gly 65 70 75 80 Pro Asp Phe Tyr Gly Ser Ser Val Ser Tyr Leu Gly Val Pro Ala His 85 90 95 Asp Leu Pro Asp Phe Asp Ile Ser Ala Tyr Phe Ser Ser Ala Ala Asp 100 105 110 Phe Ile His Arg Ala Leu Asn Thr Pro Gly Ala Lys Val Leu Val His 115 120 125 Cys Val Val Gly Val Ser Arg Ser Ala Thr Leu Val Leu Ala Tyr Leu 130 135 140 Met Leu His Gln Arg Leu Ser Leu Arg Gln Ala Val Ile Thr Val Arg 145 150 155 160 Gln His Arg Trp Val Phe Pro Asn Arg Gly Phe Leu His Gln Leu Cys 165 170 175 Arg Leu Asp His Trp Ser Leu Leu Pro Ala Met Gly Leu Cys His Phe 180 185 190 Ala Thr Leu Ala Leu Ile Leu Leu Val Leu Leu Glu Ala Leu Ala Gln 195 200 205 Ala Asp Thr Gln Lys Met Val Glu Ala Gln Arg Gly Val Gly Pro Arg 210 215 220 Ala Cys Tyr Ser Ile Trp Leu Leu Leu Ala Pro Thr Pro Pro Leu Ser 225 230 235 240 His Cys Leu Gln Ser Pro Gln Lys Gln His Gln Val Cys Gly Asp Arg 245 250 255 Arg Leu Lys Ala Ser Ser Thr Asn Cys Pro Ser Glu Lys Cys Thr Ala 260 265 270 Trp Ala Arg Tyr Ser His Arg Trp Ala His Ile Leu Val Pro Leu Lys 275 280 285 Ile Gln Leu Arg Arg Val Pro Asp Ser Phe Ser Gln Gln Met Pro Glu 290 295 300 Thr Ser Tyr Leu Thr Arg Val Gly Pro Asp Ile Gln Cys Trp Pro Glu 305 310 315 320 Ser Trp Gly Met Asp Ser Leu Gln Lys Gln Asp Leu Arg Arg Pro Lys 325 330 335 Ile His Gly Ala Val Gln Ala Ser Pro Tyr Gln Pro Pro Thr Leu Ala 340 345 350 Ser Leu Gln Arg Leu Leu Trp Val Arg Gln Ala Ala Thr Leu Asn His 355 360 365 Ile Asp Glu Val Trp Pro Ser Leu Phe Leu Gly Asp Ala Tyr Ala Ala 370 375 380 Arg Asp Lys Ser Lys Leu Ile Gln Leu Gly Ile Thr His Val Val Asn 385 390 395 400 Ala Ala Ala Gly Lys Phe Gln Val Asp Thr Gly Ala Lys Phe Tyr Arg 405 410 415 Gly Met Ser Leu Glu Tyr Tyr Gly Ile Glu Ala Asp Asp Asn Pro Phe 420 425 430 Phe Asp Leu Ser Val Tyr Phe Leu Pro Val Ala Arg Tyr Ile Arg Ala 435 440 445 Ala Leu Ser Val Pro Gln Glu Asp Gly His Gly Cys Leu Phe Phe Pro 450 455 460 Lys Gly Trp Val Val Gln Gly Gln Val Ala Asp Ala Lys Leu Val Leu 465 470 475 480 Pro Thr Gly Arg Val Leu Val His Cys Ala Met Gly Val Ser Arg Ser 485 490 495 Ala Thr Leu Val Leu Ala Phe Leu Met Ile Cys Glu Asn Met Thr Leu 500 505 510 Val Glu Ala Ile Gln Thr Val Gln Ala His Arg Asn Ile Cys Pro Asn 515 520 525 Ser Gly Phe Leu Arg Gln Leu Gln Val Leu Asp Asn Arg Leu Gly Arg 530 535 540 Glu Thr Gly Arg Phe 545 19 211 PRT Homo sapiens 19 Met Cys Pro Gly Asn Trp Leu Trp Ala Ser Met Thr Phe Met Ala Arg 1 5 10 15 Phe Ser Arg Ser Ser Ser Arg Ser Pro Val Arg Thr Arg Gly Thr Leu 20 25 30 Glu Glu Met Pro Thr Val Gln His Pro Phe Leu Asn Val Phe Glu Leu 35 40 45 Glu Arg Leu Leu Tyr Thr Gly Lys Thr Ala Cys Asn His Ala Asp Glu 50 55 60 Val Trp Pro Gly Leu Tyr Leu Gly Asp Gln Asp Met Ala Asn Asn Arg 65 70 75 80 Arg Glu Leu Arg Arg Leu Gly Ile Thr His Val Leu Asn Ala Ser His 85 90 95 Ser Arg Trp Arg Gly Thr Pro Glu Ala Tyr Glu Gly Leu Gly Ile Arg 100 105 110 Tyr Leu Gly Val Glu Ala His Asp Ser Pro Ala Phe Asp Met Ser Ile 115 120 125 His Phe Gln Thr Ala Ala Asp Phe Ile His Arg Ala Leu Ser Gln Pro 130 135 140 Gly Gly Lys Ile Leu Val His Cys Ala Val Gly Val Ser Arg Ser Ala 145 150 155 160 Thr Leu Val Leu Ala Tyr Leu Met Leu Tyr His His Leu Thr Leu Val 165 170 175 Glu Ala Ile Lys Lys Val Lys Asp His Arg Gly Ile Ile Pro Asn Arg 180 185 190 Gly Phe Leu Arg Gln Leu Leu Ala Leu Asp Arg Arg Leu Arg Gln Gly 195 200 205 Leu Glu Ala 210 20 329 PRT Homo sapiens 20 Met Gln Gly Gln Thr Val Val Pro Lys Asp Ser Tyr Thr Ile Ser Leu 1 5 10 15 Ile Gln Arg Leu Arg Gly Arg Glu Ala Ala Arg Arg Thr His Glu Asn 20 25 30 Leu Leu Arg Leu Ser Ala Leu Val Arg Ser Pro Gln Thr Ala Ser Ile 35 40 45 Asp Cys His Thr Trp Ser Val Ser Ser Gly Thr Asn Thr Ser Leu Gln 50 55 60 Ala Ser Gly Leu Gly Arg Gln Gly Ser Cys Asp Arg Ile Ala Ser Arg 65 70 75 80 Ala Ala Ser Trp Gly Cys Thr Arg Thr Ala Ala Pro Gly Ile Met Gly 85 90 95 Asn Gly Met Thr Lys Val Leu Pro Gly Leu Tyr Leu Gly Asn Phe Ile 100 105 110 Asp Ala Lys Asp Leu Asp Gln Leu Gly Arg Asn Lys Ile Thr His Ile 115 120 125 Ile Ser Ile His Glu Ser Pro Gln Pro Leu Leu Gln Asp Ile Thr Tyr 130 135 140 Leu Arg Ile Pro Val Ala Asp Thr Pro Glu Val Pro Ile Lys Lys His 145 150 155 160 Phe Lys Glu Cys Ile Asn Phe Ile His Cys Cys Arg Leu Asn Gly Gly 165 170 175 Asn Cys Leu Val His Cys Phe Ala Gly Ile Ser Arg Ser Thr Thr Ile 180 185 190 Val Thr Ala Tyr Val Met Thr Val Thr Gly Leu Gly Trp Arg Asp Val 195 200 205 Leu Glu Ala Ile Lys Ala Thr Arg Pro Ile Ala Asn Pro Asn Pro Gly 210 215 220 Phe Arg Gln Gln Leu Glu Glu Phe Gly Trp Ala Ser Ser Gln Lys Leu 225 230 235 240 Arg Arg Gln Leu Glu Glu Arg Phe Gly Glu Ser Pro Phe Arg Asp Glu 245 250 255 Glu Glu Leu Arg Ala Leu Leu Pro Leu Cys Lys Arg Cys Arg Gln Gly 260 265 270 Ser Ala Thr Ser Ala Ser Ser Ala Gly Pro His Ser Ala Ala Ser Glu 275 280 285 Gly Thr Val Gln Arg Leu Val Pro Arg Thr Pro Arg Glu Ala His Arg 290 295 300 Pro Leu Pro Leu Leu Ala Arg Val Lys Gln Thr Phe Ser Cys Leu Pro 305 310 315 320 Arg Cys Leu Ser Arg Lys Gly Gly Lys 325 21 360 PRT Homo sapiens 21 Met Ile Glu Asp Thr Met Thr Leu Leu Ser Leu Leu Gly Arg Ile Met 1 5 10 15 Arg Tyr Phe Leu Leu Arg Pro Glu Thr Leu Phe Leu Leu Cys Ile Ser 20 25 30 Leu Ala Leu Trp Ser Tyr Phe Phe His Thr Asp Glu Val Lys Thr Ile 35 40 45 Val Lys Ser Ser Arg Asp Ala Val Lys Met Val Lys Ser Lys Val Ala 50 55 60 Glu Thr Met Gln Asn Asp Arg Leu Gly Gly Leu Asp Val Leu Glu Ala 65 70 75 80 Glu Phe Ser Lys Thr Trp Glu Phe Lys Asn His Asn Val Ala Val Tyr 85 90 95 Ser Ile Gln Gly Arg Arg Asp His Met Glu Asp Arg Phe Glu Val Leu 100 105 110 Thr Asp Leu Ala Asn Lys Thr His Pro Ser Ile Phe Gly Ile Phe Asp 115 120 125 Gly His Gly Gly Glu Thr Ala Ala Glu Tyr Val Lys Ser Arg Leu Pro 130 135 140 Glu Ala Leu Lys Gln His Leu Gln Asp Tyr Glu Lys Asp Lys Glu Asn 145 150 155 160 Ser Val Leu Ser Tyr Gln Thr Ile Leu Glu Gln Gln Ile Leu Ser Ile 165 170 175 Asp Arg Glu Met Leu Glu Lys Leu Thr Val Ser Tyr Asp Glu Ala Gly 180 185 190 Thr Thr Cys Leu Ile Ala Leu Leu Ser Asp Lys Asp Leu Thr Val Ala 195 200 205 Asn Val Gly Asp Ser Arg Gly Val Leu Cys Asp Lys Asp Gly Asn Ala 210 215 220 Ile Pro Leu Ser His Asp His Lys Pro Tyr Gln Leu Lys Glu Arg Lys 225 230 235 240 Arg Ile Lys Arg Ala Gly Gly Phe Ile Ser Phe Asn Gly Ser Trp Arg 245 250 255 Val Gln Gly Ile Leu Ala Met Ser Arg Ser Leu Gly Asp Tyr Pro Leu 260 265 270 Lys Asn Leu Asn Val Val Ile Pro Asp Pro Asp Ile Leu Thr Phe Asp 275 280 285 Leu Asp Lys Leu Gln Pro Glu Phe Met Ile Leu Ala Ser Asp Gly Leu 290 295 300 Trp Asp Ala Phe Ser Asn Glu Glu Ala Val Arg Phe Ile Lys Glu Arg 305 310 315 320 Leu Asp Glu Pro His Phe Gly Ala Lys Ser Ile Val Leu Gln Ser Phe 325 330 335 Tyr Arg Gly Cys Pro Asp Asn Ile Thr Val Met Val Val Lys Phe Arg 340 345 350 Asn Ser Ser Lys Thr Glu Glu Gln 355 360 22 574 PRT Homo sapiens 22 Met Leu Ser Ala Pro Cys Cys Asp Asp Arg Arg Met Cys Val Cys Pro 1 5 10 15 Gly Pro Arg Arg Ile Gly Ile Pro Val Arg Ser Ser Ser Leu Pro Leu 20 25 30 Phe Ser Asp Ala Met Pro Ala Pro Thr Gln Leu Phe Phe Pro Leu Ile 35 40 45 Arg Asn Cys Glu Leu Ser Arg Ile Tyr Gly Thr Ala Cys Tyr Cys His 50 55 60 His Lys His Leu Cys Cys Ser Ser Ser Tyr Ile Pro Gln Ser Arg Leu 65 70 75 80 Arg Tyr Thr Pro His Pro Ala Tyr Ala Thr Phe Cys Arg Pro Lys Glu 85 90 95 Asn Trp Trp Gln Tyr Thr Gln Gly Arg Arg Tyr Ala Ser Thr Pro Gln 100 105 110 Lys Phe Tyr Leu Thr Pro Pro Gln Val Asn Ser Ile Leu Lys Ala Asn 115 120 125 Glu Tyr Ser Phe Lys Val Pro Glu Phe Asp Gly Lys Met Ser Val Leu 130 135 140 Ser Leu Asp Leu Thr Ala Ile Lys Leu Pro Ala Asn Ala Pro Ile Glu 145 150 155 160 Asp Arg Arg Ser Ala Ala Thr Cys Leu Gln Thr Arg Gly Met Leu Leu 165 170 175 Gly Val Phe Asp Gly His Ala Gly Cys Ala Trp Ser Gln Ala Val Ser 180 185 190 Glu Arg Leu Phe Tyr Tyr Ile Ala Gly Ser Leu Val Pro His Glu Thr 195 200 205 Leu Leu Glu Ile Glu Asn Ala Val Glu Ser Gly Arg Ala Leu Leu Pro 210 215 220 Ile Leu Gln Trp His Lys His Pro Asn Asp Tyr Phe Ser Lys Glu Ala 225 230 235 240 Ser Lys Leu Tyr Phe Asn Ser Leu Arg Thr Tyr Trp Gln Glu Leu Ile 245 250 255 Asp Leu Asn Thr Gly Glu Ser Thr Asp Ile Asp Val Lys Glu Ala Leu 260 265 270 Ile Asn Ala Phe Lys Arg Leu Asp Asn Asp Ile Ser Leu Glu Ala Gln 275 280 285 Val Gly Asp Pro Asn Ser Phe Leu Asn Tyr Leu Val Leu Arg Val Ala 290 295 300 Phe Ser Gly Ala Thr Ala Cys Val Ala His Val Asp Gly Val Asp Leu 305 310 315 320 His Val Ala Asn Thr Gly Asp Ser Arg Ala Met Leu Gly Val Gln Glu 325 330 335 Glu Asp Gly Ser Trp Ser Ala Val Thr Leu Ser Asn Asp His Asn Ala 340 345 350 Gln Asn Glu Arg Glu Leu Glu Arg Leu Lys Leu Glu His Pro Lys Ser 355 360 365 Glu Ala Lys Ser Val Val Lys Gln Asp Arg Leu Leu Gly Leu Leu Met 370 375 380 Pro Phe Arg Ala Phe Gly Asp Val Lys Phe Lys Trp Ser Ile Asp Leu 385 390 395 400 Gln Lys Arg Val Ile Glu Ser Gly Pro Asp Gln Leu Asn Asp Asn Glu 405 410 415 Tyr Thr Lys Phe Ile Pro Pro Asn Tyr His Thr Pro Pro Tyr Leu Thr 420 425 430 Ala Glu Pro Glu Val Thr Tyr His Arg Leu Arg Pro Gln Asp Lys Phe 435 440 445 Leu Val Leu Ala Thr Asp Gly Leu Trp Glu Thr Met His Arg Gln Asp 450 455 460 Val Val Arg Ile Val Gly Glu Tyr Leu Thr Gly Met His His Gln Gln 465 470 475 480 Pro Ile Ala Val Gly Gly Tyr Lys Val Thr Leu Gly Gln Met His Gly 485 490 495 Leu Leu Thr Glu Arg Arg Thr Lys Met Ser Ser Val Phe Glu Asp Gln 500 505 510 Asn Ala Ala Thr His Leu Ile Arg His Ala Val Gly Asn Asn Glu Phe 515 520 525 Gly Thr Val Asp His Glu Arg Leu Ser Lys Met Leu Ser Leu Pro Glu 530 535 540 Glu Leu Ala Arg Met Tyr Arg Asp Asp Ile Thr Ile Ile Val Val Gln 545 550 555 560 Phe Asn Ser His Val Val Gly Ala Tyr Gln Asn Gln Glu Lys 565 570 23 303 PRT Homo sapiens 23 Met Arg Leu Pro Ile Leu Phe Ala Ala Leu Leu Trp Phe Arg Gly Phe 1 5 10 15 Leu Ala Glu Glu Glu Ala Cys Leu Ser Leu Glu Gly Ser Pro Gly Arg 20 25 30 Glu Ser Ala Gly Pro Pro Val Asn Val Asn Ile Thr Ser Gln Gly Arg 35 40 45 Pro Thr Ser Leu Phe Leu Ser Trp Ala Ala Pro Gly Pro Gly Arg Phe 50 55 60 Thr His Ala Leu Arg Leu Thr Cys Leu Ser Pro Leu Ser Ser Pro Glu 65 70 75 80 Gly Gln Gln Leu Gln Ala His Thr Asn Ala Ser Ser Phe Lys Phe Gln 85 90 95 Asp Leu Val Ser Gly Gly Arg Tyr Gln Leu Glu Val Thr Ala Leu Arg 100 105 110 Pro Cys Gly Gln Asn Val Thr Ile Thr Leu Thr Ala Arg Thr Ala Pro 115 120 125 Ser Thr Val His Gly Leu Gln Leu His Ser Gly Ser Pro Ser Ser Leu 130 135 140 Glu Ala Ser Trp Gly Asp Ala Pro Gly Lys Gln Asp Gly Tyr Cys Leu 145 150 155 160 Leu Leu Tyr His Leu Glu Ser Gln Thr Leu Ala His Asn Ile Ser Met 165 170 175 Pro Leu Gly Thr Leu Ser Tyr Asn Phe Gly Asn Leu Leu Pro Gly Ile 180 185 190 Glu Tyr Ile Leu Glu Val Asn Thr Trp Ala Gly Asn Leu Gln Ala Thr 195 200 205 Thr Ser Leu His Gln Trp Thr Ala Pro Val Ser Pro Asp His Leu Val 210 215 220 Leu His Thr Leu Gly Thr Ser Ala Leu Gln Ala Ser Trp Asn Gly Ser 225 230 235 240 Lys Gly Ala Ala Trp Leu His Leu Val Leu Thr Asp Leu Leu Gly Gly 245 250 255 Thr Asn Leu Thr Ala Val Phe Arg Arg Gly Val Ser His His Thr Ser 260 265 270 Leu His Leu Ser Gln Gly Pro Pro Tyr Glu Leu Thr Leu Ser Ala Ala 275 280 285 Ala Arg Pro His Arg Ala Val Gly Pro Asn Ala Thr Glu Trp Thr 290 295 300 24 118 PRT Homo sapiens MOD_RES (1)..(118) “Xaa” represents any, other or unknown amino acid 24 Ser Glu Gly Val Gly Trp Thr Gly Cys Phe Ile Val Ile Asp Ala Met 1 5 10 15 Leu Glu Arg Ile Lys His Glu Lys Thr Val Gly Asn Tyr Ala Tyr Ala 20 25 30 Thr Leu Met Arg Thr Gln Arg Asn Tyr Met Val Gln Ala Gly Asp Gln 35 40 45 Cys Ile Ser Val His Asp Ala Leu Leu Glu Ala Val Thr Cys Val Asn 50 55 60 Thr Lys Val Pro Ala Arg Asn Leu Tyr Ala Tyr Ile Xaa Lys Leu Thr 65 70 75 80 Gln Ile Glu Arg Gly Gln Asn Val Ile Gly Val Val Leu Lys Phe Lys 85 90 95 His Leu Ile Ser Ser Lys Ala His Ile Ser Gly Phe Leu Ser Ala Asn 100 105 110 Leu Pro Cys Asn Asn Phe 115 25 12 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 25 Ser Glu Phe Leu Asp Glu Ala Leu Leu Thr Tyr Arg 1 5 10 26 34 DNA Artificial Sequence Description of Artificial Sequence Primer 26 ctgagttcct ggatgaggcg ctgctgactt acag 34 27 163 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 27 Ala Asn Gly Asn Ser Val Arg Ser Thr Ser Arg Phe Ser Ser Ser Ser 1 5 10 15 Thr Arg Glu Gly Arg Glu Met His Lys Phe Ser Arg Ser Thr Tyr Asn 20 25 30 Glu Thr Ser Ser Ser Arg Glu Glu Ser Pro Glu Pro Tyr Phe Phe Arg 35 40 45 Arg Thr Pro Glu Ser Ser Glu Arg Glu Glu Ser Pro Glu Pro Gln Arg 50 55 60 Pro Asn Trp Ala Arg Ser Arg Asp Trp Glu Asp Val Glu Glu Ser Ser 65 70 75 80 Lys Ser Asp Phe Ser Glu Phe Gly Ala Lys Arg Lys Phe Thr Gln Ser 85 90 95 Phe Met Arg Ser Glu Glu Glu Gly Glu Lys Glu Arg Thr Glu Asn Arg 100 105 110 Glu Glu Gly Arg Phe Ala Ser Gly Arg Arg Ser Gln Tyr Arg Arg Ser 115 120 125 Asn Asp Arg Glu Glu Glu Glu Glu Met Asp Asp Glu Ala Ile Ile Ala 130 135 140 Ala Trp Arg Arg Arg Gln Glu Glu Thr Arg Thr Lys Leu Gln Lys Arg 145 150 155 160 Arg Glu Asp 28 79 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 28 Gln Gly Phe Gln Val Ala Leu Glu Asn His Phe Ala Ser Arg Cys His 1 5 10 15 Arg Ser Pro Leu Arg Leu Asp Gly Thr Gln Gly Gln Arg His Arg Ser 20 25 30 Asp Pro Lys Asn Arg Pro Leu Leu Pro Val Gly Val Ala Gly Arg Ala 35 40 45 Ala Ala Thr Cys Pro Thr Asp Gln Asn Leu Pro Thr Arg Pro Gly Pro 50 55 60 Gly Thr Pro Ser Cys Arg Lys Glu Arg Lys Ser Leu Gly Ser Lys 65 70 75 29 26 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 29 Ser Gly Ser Ser Arg Phe Trp Thr Thr Asp Trp Gly Gly Arg Arg Gly 1 5 10 15 Gly Ser Asp Leu Ala Gly Ser Gln Asp Pro 20 25 30 57 DNA Artificial Sequence Description of Artificial Sequence Primer 30 aagcagtggt aacaacgcag agtacttttt tttttttttt tttttttttt tttttvn 57 31 30 DNA Artificial Sequence Description of Artificial Sequence Primer 31 aagcagtggt aacaacgcag agtacgcggg 30 32 30 DNA Artificial Sequence Description of Artificial Sequence Primer 32 aagtggcaac agagataacg cgtacgcggg 30 33 23 DNA Artificial Sequence Description of Artificial Sequence Primer 33 aagcagtggt aacaacgcag agt 23 34 23 DNA Artificial Sequence Description of Artificial Sequence Primer 34 aagtggcaac agagataacg cgt 23 35 20 DNA Artificial Sequence Description of Artificial Sequence Primer 35 ggagctgtcg tattccagtc 20 36 21 DNA Artificial Sequence Description of Artificial Sequence Primer 36 aacccctcaa gacccgttta g 21 37 5 PRT Homo sapiens 37 Met Thr Pro Glu Lys 1 5 38 18 DNA Homo sapiens 38 ccaaacataa gtggcaca 18 39 17 DNA Homo sapiens 39 agaagatgtc tgagtac 17 40 15 DNA Homo sapiens 40 catctacccc aatga 15 41 20 DNA Homo sapiens 41 cagcagcctc cgagggaacc 20 42 22 PRT Homo sapiens 42 Gly Phe Ser Val Ser Thr Ala Gly Arg Met His Ile Phe Lys Pro Val 1 5 10 15 Ser Val Gln Ala Met Trp 20 43 18 DNA Homo sapiens 43 ccaaacataa gtggcaca 18 44 13 PRT Homo sapiens 44 Tyr Cys His Tyr Ile Ile Phe Ser Cys Val Phe Ile Ser 1 5 10 45 37 DNA Homo sapiens 45 actgtcatta catcattttc tcttgtgttt tcatttc 37 46 4 PRT Homo sapiens 46 Val His Leu Leu 1 47 6 PRT Homo sapiens 47 Met Leu Glu Ser Ala Glu 1 5 48 17 DNA Homo sapiens 48 agaagatgtc tgagtac 17 49 15 DNA Homo sapiens 49 catctacccc aatga 15 50 240 DNA Homo sapiens 50 caagggtttc aggtcgcact ggaaaatcat tttgcaagca gatgtcatag gtctcctctt 60 agactggacg gcacgcaagg tcagcgtcac agatctgacc ctaaaaatag gcctctgttg 120 ccagtcgggg tggctgggcg tgcggctgct acatgcccca cggaccagaa cctcccgacg 180 cggccaggcc ccggcacacc cagctgcaga aaggagagaa aatcccttgg ctctaaaatg 240 51 50 PRT Homo sapiens 51 Met Ala Glu Thr Ser Leu Pro Glu Leu Gly Gly Glu Asp Lys Ala Thr 1 5 10 15 Pro Cys Pro Ser Ile Leu Glu Leu Glu Glu Leu Leu Arg Ala Gly Lys 20 25 30 Ser Ser Cys Ser Arg Val Asp Glu Val Trp Pro Asn Leu Phe Ile Gly 35 40 45 Asp Ala 50 52 73 PRT Homo sapiens 52 Ala Thr Ala Asn Asn Arg Phe Glu Leu Trp Lys Leu Gly Ile Thr His 1 5 10 15 Val Leu Asn Ala Ala His Lys Gly Leu Tyr Cys Gln Gly Gly Pro Asp 20 25 30 Phe Tyr Gly Ser Ser Val Ser Tyr Leu Gly Val Pro Ala His Asp Leu 35 40 45 Pro Asp Phe Asp Ile Ser Ala Tyr Phe Ser Ser Ala Ala Asp Phe Ile 50 55 60 His Arg Ala Leu Asn Thr Pro Gly Ala 65 70 53 57 PRT Homo sapiens 53 Lys Val Leu Val His Cys Val Val Gly Val Ser Arg Ser Ala Thr Leu 1 5 10 15 Val Leu Ala Tyr Leu Met Leu His Gln Arg Leu Ser Leu Arg Gln Ala 20 25 30 Val Ile Thr Val Arg Gln His Arg Trp Val Phe Pro Asn Arg Gly Phe 35 40 45 Leu His Gln Leu Cys Arg Leu Asp His 50 55 54 67 PRT Homo sapiens 54 Trp Ser Leu Leu Pro Ala Met Gly Leu Cys His Phe Ala Thr Leu Ala 1 5 10 15 Leu Ile Leu Leu Val Leu Leu Glu Ala Leu Ala Gln Ala Asp Thr Gln 20 25 30 Lys Met Val Glu Ala Gln Arg Gly Val Gly Pro Arg Ala Cys Tyr Ser 35 40 45 Ile Trp Leu Leu Leu Ala Pro Thr Pro Pro Leu Ser His Cys Leu Gln 50 55 60 Ser Pro Gln 65 55 33 PRT Homo sapiens 55 Lys Gln His Gln Val Cys Gly Asp Arg Arg Leu Lys Ala Ser Ser Thr 1 5 10 15 Asn Cys Pro Ser Glu Lys Cys Thr Ala Trp Ala Arg Tyr Ser His Arg 20 25 30 Trp 56 43 PRT Homo sapiens 56 Ala His Ile Leu Val Pro Leu Lys Ile Gln Leu Arg Arg Val Pro Asp 1 5 10 15 Ser Phe Ser Gln Gln Met Pro Glu Thr Ser Tyr Leu Thr Arg Val Gly 20 25 30 Pro Asp Ile Gln Cys Trp Pro Glu Ser Trp Gly 35 40 57 58 PRT Homo sapiens 57 Met Asp Ser Leu Gln Lys Gln Asp Leu Arg Arg Pro Lys Ile His Gly 1 5 10 15 Ala Val Gln Ala Ser Pro Tyr Gln Pro Pro Thr Leu Ala Ser Leu Gln 20 25 30 Arg Leu Leu Trp Val Arg Gln Ala Ala Thr Leu Asn His Ile Asp Glu 35 40 45 Val Trp Pro Ser Leu Phe Leu Gly Asp Ala 50 55 58 74 PRT Homo sapiens 58 Tyr Ala Ala Arg Asp Lys Ser Lys Leu Ile Gln Leu Gly Ile Thr His 1 5 10 15 Val Val Asn Ala Ala Ala Gly Lys Phe Gln Val Asp Thr Gly Ala Lys 20 25 30 Phe Tyr Arg Gly Met Ser Leu Glu Tyr Tyr Gly Ile Glu Ala Asp Asp 35 40 45 Asn Pro Phe Phe Asp Leu Ser Val Tyr Phe Leu Pro Val Ala Arg Tyr 50 55 60 Ile Arg Ala Ala Leu Ser Val Pro Gln Glu 65 70 59 94 PRT Homo sapiens 59 Asp Gly His Gly Cys Leu Phe Phe Pro Lys Gly Trp Val Val Gln Gly 1 5 10 15 Gln Val Ala Asp Ala Lys Leu Val Leu Pro Thr Gly Arg Val Leu Val 20 25 30 His Cys Ala Met Gly Val Ser Arg Ser Ala Thr Leu Val Leu Ala Phe 35 40 45 Leu Met Ile Cys Glu Asn Met Thr Leu Val Glu Ala Ile Gln Thr Val 50 55 60 Gln Ala His Arg Asn Ile Cys Pro Asn Ser Gly Phe Leu Arg Gln Leu 65 70 75 80 Gln Val Leu Asp Asn Arg Leu Gly Arg Glu Thr Gly Arg Phe 85 90 60 4 PRT Homo sapiens 60 Asn Ser Gly Phe 1 61 4 PRT Homo sapiens 61 Met Gly Asn Gly 1 62 20 DNA Homo sapiens 62 cagcagcctc cgagggaacc 20 63 19 DNA Homo sapiens 63 ccaaacataa gtggcacar 19 64 16 DNA Homo sapiens 64 catctacccc aatgas 16 65 18 DNA Homo sapiens 65 agaagatgtc tgagtacm 18 66 21 DNA Homo sapiens 66 cagcagcctc cgagggaaccs 21 67 522 DNA Homo sapiens 67 gattccgcag ccaagtccag acaaggcagt ggtgccaagc ggcagctgga tgggctggag 60 gcctccaagg agcccgggag acgggccctg ctctacacag agggaaaccc gggcctcctt 120 ggaaacatct ctgtgccacc tggtgccacc cacatcacct tctatgggcc agtgcctggg 180 gcccgctact gtgtggacat tgcctcatct ctgggaatca tcacttacag cctcatgggc 240 cacaaaagtc ccctggcacc acagtccctg gaggttatca gcaggggtgg cccctctgac 300 ctggccattg tctgggcccc agcaccagga cagcgggaag gctacagggt cgcttggcac 360 caggagggca gccagaggtc accgggcagt gcttgttgat tgggcccgga caattccagc 420 ctgactctga ggagtctggt gcccggctcc tcctatgcca tgtcagtgtg ggcctgggca 480 gagaaccttg gctctagcat ccagaagatc cacccctgta ct 522 68 1692 DNA Homo sapiens modified_base (1)..(1692) “n” represents a, t, c, g, other or unknown 68 ccgcttgccc ctcctctggt aaatgtgacg agtgaaggtc ccacccagct ctgggcatcc 60 tgggtccatg cccccagggg ccgagacagc tacccggtga ccctgtaccg ggcaggcacc 120 agcgccgtcg gagccaaggt ggccagcaca agcttttcaa gtctgactcc aggcacgaag 180 tacaaggtgg aggttgtcac gcaggctggg ccccaccaca ttgcagcagc caacacctct 240 ggctggaccc atgaggcatg gggggaaggc agcgatgcag gaaaagccct gcacacaccc 300 agtgagttgg tgtccatgca tgcgagcacc gctgtggtca acctggcctg ggccagcagc 360 cccttggggc aggggatgtg ctacacccaa ctctcagagg cggggcacct ctcctgggag 420 caccctctgg tgccaggcca agcccacctc atcctgaggg gcctcacacc tggatgcaac 480 ctctccctgt cagtgctgtg ccaggcaggg ccgctgcagg cgtccactca gcgcgtggta 540 ctgcttgttg agcctggccc tgtggaagat gtgcagtgcc agcctgaggc caccttcctg 600 gccctgaact ggacagtgcc cgccagggat gtgggcacct gtctggtggt ggcagagcag 660 ctggtggcag gagggaatgc tcaccttgtg ttccaggccg acacctccaa aaatgcagtc 720 ctgttgccca acctggtgcc tgtcacttcc tatcacctca gcctcgccgt gctgggcagg 780 aacggtctgt ggagtcgggt ggtcactctg gcatgttcca catctgccga ggcctggcat 840 cccccagcgt tagccccggc ccctgagctg gagcctggga cagaaatggg agtgatgatc 900 ccgcggggta tgtttggcaa ggatgatggg cagatccagt ggtacggcat cattgccacc 960 accaacatgt cactgcctca gccttcctgg gaagccatca accacatgtg gcatgaccac 1020 tactacagag gacatgactc ctacctggcc atcctgctcc ccaacccctt ctacccggat 1080 ccctgggctg tgccgagatc ctggacagtg cctgtgggta cagaggactg tggccacacc 1140 aaagagatat gcaacgggca gctcaagcta ggtcctgttt ctctgcccag gttcagcgtt 1200 gcagccttta ccaggtacag ccctcctgag accattaact ccttctcagc cttctcgnag 1260 ccctgggccg gtgtctccct ggcatcagtg cccctgccgg taatggaggg cctcgtggtg 1320 ggctgtgtcc tcaccatctg tgctgtgctg ggcctgctgt gctggaggcg ggtgaagggg 1380 cagagggcag ggaagaatcc attttcccaa gagctgacag cttacaacct gcggtagacc 1440 caccggccca tccctatcca cagcttcagg cagagctatg aggccaagag tgctcacgca 1500 caccaggctt tctttttgca attcgaggag ctgaaggagg tgggcaagga gcagcccaga 1560 ctggaggctg agtacgctgc caacaccacc aagaaccatt acccacatgt gcttccctac 1620 gaccactcca gggtcaggct gacccagctg gagggagagc ctcattctga ctacatcaat 1680 gccaacttca tc 1692 69 333 DNA Homo sapiens 69 gctacaccca cccacccaca ggaattcatt gcctctcagg cgcctctcaa gaaaacgctg 60 gagaacttct ggcggctggt gcgggagtag caggtccgca tcatcatcat gctgaccgtc 120 ggcatggaga acaggagggt gctgtgtgag cattactggc tgaccgactc taccccggtc 180 acccatgatc acatcaccat ccacctccta gccgaggagg ctgacgatga gtggaccaag 240 cgggaattcc agctgcagca catgcgtgcc ccaaggatga gggggttgtc cagcaacagc 300 agcggagggt ggagtaactg caattcacca cct 333 70 1191 DNA Homo sapiens 70 cctgaccaca gcatcctcaa ggcccccagc tccctgctta cctttatgga gctggtacag 60 gaacaggcaa gggccaccca gggcatggga cccatcctgg tgcactgcag gagggcagtg 120 tgggcatgga ggcagacggg caccttcgtg gccctgttga ggctgctgca gcagctggag 180 gaggagcaga tggtagatgt gttccatgct gtgtttgcat tctggatgca cgggcccctc 240 atgatccaga ccctgagcca gtacgtcttc ctgcacagct gcctactgaa caagattctg 300 gaagggccct tcaacatctc tgagtcttgg cccatctctg tgatgaactt cgcacaggcg 360 tgtgccaaga gggcagccaa tgccaacgct ggcttcttga aggagtacga gctcttgctg 420 caggccatca aggacgaggc tggctcttac gcacccctgc ctggctatga gcaggacagc 480 cccatctcct gtgagtctca ctgggacacc ctcagtctct ggaagccaat gagctgtgct 540 ctgcagggtg ggccctctgg ctgtgatcat atggtgctga ctggcctcgc agggccagag 600 gagctctggg agctggtgtg gcagcacggg gctcatgtgc ttgtctctct gtgcccactc 660 gatgccatgg agaagccaca ggaattctgg ccaatggaga tgcagcccat agtcacagac 720 atggtgacag tgcactgggt ggccgagagc agcacagtgg gctggctctg tgccctcttc 780 agggtcacac atgtagcacc aatgccgatc atgtctttgc ccgaggggga gagtaggaag 840 gaaagggagg tgcagagact gcagtttcca tacctggagc ctgggcatga gctgcccgcc 900 accaccctgc tgcccttcct ggctgctgtg ggccagtgct gctctcgggg caacagcaag 960 aagccgggca cactgctcag ccactccagc aagggtgcga cccagctggg caccttcctg 1020 gccatggagc agctgctgca gcaggcaggg tctgagtgca ccgtggatgt ctttaacgtg 1080 gccctgcagc agtctcaggc ctgtgacctt atgaccccaa cgctgaagca gtatatctac 1140 ctctacaatt gtctgaacag cgcactggca gacgggctgc ccctgagtcg g 1191 71 57 DNA Homo sapiens 71 cactggtcac tgtgcaggag aggtttgtgc cctgtgggat ggggacagca ttcctga 57 72 174 PRT Homo sapiens 72 Asp Ser Ala Ala Lys Ser Arg Gln Gly Ser Gly Ala Lys Arg Gln Leu 1 5 10 15 Asp Gly Leu Glu Ala Ser Lys Glu Pro Gly Arg Arg Ala Leu Leu Tyr 20 25 30 Thr Glu Gly Asn Pro Gly Leu Leu Gly Asn Ile Ser Val Pro Pro Gly 35 40 45 Ala Thr His Ile Thr Phe Tyr Gly Pro Val Pro Gly Ala Arg Tyr Cys 50 55 60 Val Asp Ile Ala Ser Ser Leu Gly Ile Ile Thr Tyr Ser Leu Met Gly 65 70 75 80 His Lys Ser Pro Leu Ala Pro Gln Ser Leu Glu Val Ile Ser Arg Gly 85 90 95 Gly Pro Ser Asp Leu Ala Ile Val Trp Ala Pro Ala Pro Gly Gln Arg 100 105 110 Glu Gly Tyr Arg Val Ala Trp His Gln Glu Gly Ser Gln Arg Ser Pro 115 120 125 Gly Ser Leu Val Asp Leu Gly Pro Asp Asn Ser Ser Leu Thr Leu Arg 130 135 140 Ser Leu Val Pro Gly Ser Ser Tyr Ala Met Ser Val Trp Ala Trp Ala 145 150 155 160 Glu Asn Leu Gly Ser Ser Ile Gln Lys Ile His Pro Cys Thr 165 170 73 563 PRT Homo sapiens MOD_RES (1)..(563) “Xaa” represents any, other or unknown amino acid 73 Pro Leu Ala Pro Pro Leu Val Asn Val Thr Ser Glu Gly Pro Thr Gln 1 5 10 15 Leu Trp Ala Ser Trp Val His Ala Pro Arg Gly Arg Asp Ser Tyr Pro 20 25 30 Val Thr Leu Tyr Arg Ala Gly Thr Ser Ala Val Gly Ala Lys Val Ala 35 40 45 Ser Thr Ser Phe Ser Ser Leu Thr Pro Gly Thr Lys Tyr Lys Val Glu 50 55 60 Val Val Thr Gln Ala Gly Pro His His Ile Ala Ala Ala Asn Thr Ser 65 70 75 80 Gly Trp Thr His Glu Ala Trp Gly Glu Gly Ser Asp Ala Gly Lys Ala 85 90 95 Leu His Thr Pro Ser Glu Leu Val Ser Met His Ala Ser Thr Ala Val 100 105 110 Val Asn Leu Ala Trp Ala Ser Ser Pro Leu Gly Gln Gly Met Cys Tyr 115 120 125 Thr Gln Leu Ser Glu Ala Gly His Leu Ser Trp Glu His Pro Leu Val 130 135 140 Pro Gly Gln Ala His Leu Ile Leu Arg Gly Leu Thr Pro Gly Cys Asn 145 150 155 160 Leu Ser Leu Ser Val Leu Cys Gln Ala Gly Pro Leu Gln Ala Ser Thr 165 170 175 Gln Arg Val Val Leu Leu Val Glu Pro Gly Pro Val Glu Asp Val Gln 180 185 190 Cys Gln Pro Glu Ala Thr Phe Leu Ala Leu Asn Trp Thr Val Pro Ala 195 200 205 Arg Asp Val Gly Thr Cys Leu Val Val Ala Glu Gln Leu Val Ala Gly 210 215 220 Gly Asn Ala His Leu Val Phe Gln Ala Asp Thr Ser Lys Asn Ala Val 225 230 235 240 Leu Leu Pro Asn Leu Val Pro Val Thr Ser Tyr His Leu Ser Leu Ala 245 250 255 Val Leu Gly Arg Asn Gly Leu Trp Ser Arg Val Val Thr Leu Ala Cys 260 265 270 Ser Thr Ser Ala Glu Ala Trp His Pro Pro Ala Leu Ala Pro Ala Pro 275 280 285 Glu Leu Glu Pro Gly Thr Glu Met Gly Val Met Ile Pro Arg Gly Met 290 295 300 Phe Gly Lys Asp Asp Gly Gln Ile Gln Trp Tyr Gly Ile Ile Ala Thr 305 310 315 320 Thr Asn Met Ser Leu Pro Gln Pro Ser Trp Glu Ala Ile Asn His Met 325 330 335 Trp His Asp His Tyr Tyr Arg Gly His Asp Ser Tyr Leu Ala Ile Leu 340 345 350 Leu Pro Asn Pro Phe Tyr Pro Asp Pro Trp Ala Val Pro Arg Ser Trp 355 360 365 Thr Val Pro Val Gly Thr Glu Asp Cys Gly His Thr Lys Glu Ile Cys 370 375 380 Asn Gly Gln Leu Lys Leu Gly Pro Val Ser Leu Pro Arg Phe Ser Val 385 390 395 400 Ala Ala Phe Thr Arg Tyr Ser Pro Pro Glu Thr Ile Asn Ser Phe Ser 405 410 415 Ala Phe Ser Xaa Pro Trp Ala Gly Val Ser Leu Ala Ser Val Pro Leu 420 425 430 Pro Val Met Glu Gly Leu Val Val Gly Cys Val Leu Thr Ile Cys Ala 435 440 445 Val Leu Gly Leu Leu Cys Trp Arg Arg Val Lys Gly Gln Arg Ala Gly 450 455 460 Lys Asn Pro Phe Ser Gln Glu Leu Thr Ala Tyr Asn Leu Arg Thr His 465 470 475 480 Arg Pro Ile Pro Ile His Ser Phe Arg Gln Ser Tyr Glu Ala Lys Ser 485 490 495 Ala His Ala His Gln Ala Phe Phe Leu Gln Phe Glu Glu Leu Lys Glu 500 505 510 Val Gly Lys Glu Gln Pro Arg Leu Glu Ala Glu Tyr Ala Ala Asn Thr 515 520 525 Thr Lys Asn His Tyr Pro His Val Leu Pro Tyr Asp His Ser Arg Val 530 535 540 Arg Leu Thr Gln Leu Glu Gly Glu Pro His Ser Asp Tyr Ile Asn Ala 545 550 555 560 Asn Phe Ile 74 110 PRT Homo sapiens 74 Ala Thr Pro Thr His Pro Gln Glu Phe Ile Ala Ser Gln Ala Pro Leu 1 5 10 15 Lys Lys Thr Leu Glu Asn Phe Trp Arg Leu Val Arg Glu Gln Val Arg 20 25 30 Ile Ile Ile Met Leu Thr Val Gly Met Glu Asn Arg Arg Val Leu Cys 35 40 45 Glu His Tyr Trp Leu Thr Asp Ser Thr Pro Val Thr His Asp His Ile 50 55 60 Thr Ile His Leu Leu Ala Glu Glu Ala Asp Asp Glu Trp Thr Lys Arg 65 70 75 80 Glu Phe Gln Leu Gln His Met Arg Ala Pro Arg Met Arg Gly Leu Ser 85 90 95 Ser Asn Ser Ser Gly Gly Trp Ser Asn Cys Asn Ser Pro Pro 100 105 110 75 397 PRT Homo sapiens 75 Pro Asp His Ser Ile Leu Lys Ala Pro Ser Ser Leu Leu Thr Phe Met 1 5 10 15 Glu Leu Val Gln Glu Gln Ala Arg Ala Thr Gln Gly Met Gly Pro Ile 20 25 30 Leu Val His Cys Arg Arg Ala Val Trp Ala Trp Arg Gln Thr Gly Thr 35 40 45 Phe Val Ala Leu Leu Arg Leu Leu Gln Gln Leu Glu Glu Glu Gln Met 50 55 60 Val Asp Val Phe His Ala Val Phe Ala Phe Trp Met His Gly Pro Leu 65 70 75 80 Met Ile Gln Thr Leu Ser Gln Tyr Val Phe Leu His Ser Cys Leu Leu 85 90 95 Asn Lys Ile Leu Glu Gly Pro Phe Asn Ile Ser Glu Ser Trp Pro Ile 100 105 110 Ser Val Met Asn Phe Ala Gln Ala Cys Ala Lys Arg Ala Ala Asn Ala 115 120 125 Asn Ala Gly Phe Leu Lys Glu Tyr Glu Leu Leu Leu Gln Ala Ile Lys 130 135 140 Asp Glu Ala Gly Ser Tyr Ala Pro Leu Pro Gly Tyr Glu Gln Asp Ser 145 150 155 160 Pro Ile Ser Cys Glu Ser His Trp Asp Thr Leu Ser Leu Trp Lys Pro 165 170 175 Met Ser Cys Ala Leu Gln Gly Gly Pro Ser Gly Cys Asp His Met Val 180 185 190 Leu Thr Gly Leu Ala Gly Pro Glu Glu Leu Trp Glu Leu Val Trp Gln 195 200 205 His Gly Ala His Val Leu Val Ser Leu Cys Pro Leu Asp Ala Met Glu 210 215 220 Lys Pro Gln Glu Phe Trp Pro Met Glu Met Gln Pro Ile Val Thr Asp 225 230 235 240 Met Val Thr Val His Trp Val Ala Glu Ser Ser Thr Val Gly Trp Leu 245 250 255 Cys Ala Leu Phe Arg Val Thr His Val Ala Pro Met Pro Ile Met Ser 260 265 270 Leu Pro Glu Gly Glu Ser Arg Lys Glu Arg Glu Val Gln Arg Leu Gln 275 280 285 Phe Pro Tyr Leu Glu Pro Gly His Glu Leu Pro Ala Thr Thr Leu Leu 290 295 300 Pro Phe Leu Ala Ala Val Gly Gln Cys Cys Ser Arg Gly Asn Ser Lys 305 310 315 320 Lys Pro Gly Thr Leu Leu Ser His Ser Ser Lys Gly Ala Thr Gln Leu 325 330 335 Gly Thr Phe Leu Ala Met Glu Gln Leu Leu Gln Gln Ala Gly Ser Glu 340 345 350 Cys Thr Val Asp Val Phe Asn Val Ala Leu Gln Gln Ser Gln Ala Cys 355 360 365 Asp Leu Met Thr Pro Thr Leu Lys Gln Tyr Ile Tyr Leu Tyr Asn Cys 370 375 380 Leu Asn Ser Ala Leu Ala Asp Gly Leu Pro Leu Ser Arg 385 390 395 76 18 PRT Homo sapiens 76 His Trp Ser Leu Cys Arg Arg Gly Leu Cys Pro Val Gly Trp Gly Gln 1 5 10 15 His Ser

Claims (29)

What is claimed is:
1. An isolated, enriched or purified nucleic acid molecule encoding a phosphatase polypeptide, wherein said nucleic acid molecule comprises a nucleotide sequence that:
(a) encodes a polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ D NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ED NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24;
(b) is the complement of the nucleotide sequence of (a);
(c) hybridizes under stringent conditions to the nucleotide molecule of (a) and encodes a naturally occurring phosphatase polypeptide;
(d) encodes a polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ D NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ED NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, except that said polypeptide lacks one or more, but not all, of an N-terminal domain, a C-terminal catalytic domain, a catalytic domain, a C-terminal domain, a coiled-coil structure region, a proline-rich region, a spacer region and a C-terminal tail; or
(e) is the complement of the nucleotide sequence of (d).
2. The nucleic acid molecule of claim 1, further comprising a vector or promoter effective to initiate transcription in a host cell.
3. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule is isolated, enriched, or purified from a mammal.
4. The nucleic acid molecule of claim 3, wherein said mammal is a human.
5. The nucleic acid probe of claim 1 used for the detection of nucleic acid encoding a phosphatase polypeptide in a sample, wherein said phosphatase polypeptide is selected from the group consisting of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ D NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
6. A recombinant cell comprising the nucleic acid molecule of claim 1 encoding a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
7. An isolated, enriched, or purified phosphatase polypeptide, wherein said polypeptide comprises an amino acid sequence having
(a) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24; or
(b) an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, except that the polypeptide lacks one or more, but not all, of the domains selected from the group consisting of an N-terminal domain, a C-terminal catalytic domain, a catalytic domain, a C-terminal domain, a coiled-coil structure region, a proline-rich region, a spacer region, and a C-terminal tail.
8. The phosphatase polypeptide of claim 7, wherein said polypeptide is isolated, purified, or enriched from a mammal.
9. The phosphatase polypeptide of claim 8, wherein said mammal is a human.
10. An antibody or antibody fragment having specific binding affinity to a phosphatase polypeptide or to a domain of said polypeptide, wherein said polypeptide is a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
11. A hybridoma which produces an antibody having specific binding affinity to a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
12. A kit comprising an antibody which binds to a polypeptide of claim 7 or 8 and negative control antibody.
13. A method for identifying a substance that modulates the activity of a phosphatase polypeptide comprising the steps of:
(a) contacting the phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24 with a test substance;
(b) measuring the activity of said polypeptide; and
(c) determining whether said substance modulates the activity of said polypeptide.
14. A method for identifying a substance that modulates the activity of a phosphatase polypeptide in a cell comprising the steps of:
(a) expressing a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ]D NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24;
(b) adding a test substance to said cell; and
(c) monitoring a change in cell phenotype or the interaction between said polypeptide and a natural binding partner.
15. A method for treating a disease or disorder by administering to a patient in need of such treatment a substance that modulates the activity of a phosphatase having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24.
16. The method of claim 15, wherein said disease or disorder is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
17. The method of claim 15, wherein said disease or disorder is selected from the group consisting of cancers of tissues; cancers of hematopoietic origin; diseases of the central nervous system; diseases of the peripheral nervous system; Alzheimer's disease; Parkinson's disease; multiple sclerosis; amyotrophic lateral sclerosis; viral infections; infections caused by prions; infections caused by bacteria; infections caused by fungi; ocular diseases, metabolic disorders, and diabetes.
18. The method of claim 15, wherein said disease or disorder is selected from the group consisting of migraines; pain; sexual dysfunction; mood disorders; attention disorders; cognition disorders; hypotension; hypertension; psychotic disorders; neurological disorders; dyskinesias; metabolic disorders; and organ transplant rejection.
19. The method of claim 15, wherein said substance modulates phosphatase activity in vitro.
20. The method of claim 19, wherein said substance is a phosphatase inhibitor.
21. A method for detection of a phosphatase polypeptide in a sample as a diagnostic tool for a disease or disorder, wherein said method comprises:
(a) contacting said sample with a nucleic acid probe which hybridizes under hybridization assay conditions to a nucleic acid target region of a phosphatase polypeptide having an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, said probe comprising the nucleic acid sequence encoding said polypeptide, fragments thereof, or the complements of said sequences and fragments; and
(b) detecting the presence or amount of the probe: target region hybrid as an indication of said disease.
22. The method of claim 21, wherein said disease or disorder is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
23. The method of claim 21, wherein said disease or disorder is selected from the group consisting of cancers of tissues; cancers of hematopoietic origin; diseases of the central nervous system; diseases of the peripheral nervous system; Alzheimer's disease; Parkinson's disease; multiple sclerosis; amyotrophic lateral sclerosis; viral infections; infections caused by prions; infections caused by bacteria; infections caused by fungi; and ocular diseases.
24. The method of claim 21, wherein said disease or disorder is selected from the group consisting of migraines, pain; sexual dysfunction; mood disorders; attention disorders; cognition disorders; hypotension; hypertension; psychotic disorders; neurological disorders; dyskinesias; metabolic disorders; and organ transplant rejection.
25. A method for detection of a phosphatase polypeptide in a sample as a diagnostic tool for a disease or disorder, wherein said method comprises:
(a) comparing a nucleic acid target region encoding said phosphatase polypeptide in a sample, wherein said phosphatase polypeptide has an amino acid sequence selected from the group consisting of those set forth in SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, or one or more fragments thereof, with a control nucleic acid target region encoding said phosphatase polypeptide, or one or more fragments thereof; and
(b) detecting differences in sequence or amount between said target region and said control target region, as an indication of said disease or disorder.
26. The method of claim 25, wherein said disease or disorder is selected from the group consisting of cancers, immune-related diseases and disorders, cardiovascular disease, brain or neuronal-associated diseases, and metabolic disorders.
27. The method of claim 25, wherein said disease or disorder is selected from the group consisting of cancers of tissues; cancers of hematopoietic origin; diseases of the central nervous system; diseases of the peripheral nervous system; Alzheimer's disease; Parkinson's disease; multiple sclerosis; amyotrophic lateral sclerosis; viral infections; infections caused by prions; infections caused by bacteria; infections caused by fungi; and ocular diseases.
28. The method of claim 25, wherein said disease or disorder is selected from the group consisting of migraines, pain; sexual dysfunction; mood disorders; attention disorders; cognition disorders; hypotension; hypertension; psychotic disorders; neurological disorders; dyskinesias; metabolic disorders; and organ transplant rejection.
29. A nucleic acid that encodes a mammalian phosphatase or a fragment thereof selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12.
US10/168,506 1999-12-21 2000-12-21 Mammalian protein phosphatases Abandoned US20040053229A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/168,506 US20040053229A1 (en) 2000-12-21 2000-12-21 Mammalian protein phosphatases
US10/838,181 US20050084877A1 (en) 1999-12-21 2004-05-04 Mammalian protein phosphatases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/168,506 US20040053229A1 (en) 2000-12-21 2000-12-21 Mammalian protein phosphatases
PCT/US2000/034736 WO2001046394A2 (en) 1999-12-21 2000-12-21 Mammalian protein phosphatases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/838,181 Division US20050084877A1 (en) 1999-12-21 2004-05-04 Mammalian protein phosphatases

Publications (1)

Publication Number Publication Date
US20040053229A1 true US20040053229A1 (en) 2004-03-18

Family

ID=31990153

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/168,506 Abandoned US20040053229A1 (en) 1999-12-21 2000-12-21 Mammalian protein phosphatases
US10/838,181 Abandoned US20050084877A1 (en) 1999-12-21 2004-05-04 Mammalian protein phosphatases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/838,181 Abandoned US20050084877A1 (en) 1999-12-21 2004-05-04 Mammalian protein phosphatases

Country Status (1)

Country Link
US (2) US20040053229A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005067649A2 (en) * 2004-01-08 2005-07-28 The Ohio State University Use of databases to create gene expression microarrays

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100190697A1 (en) * 2006-09-13 2010-07-29 The Trustees Of Columbia University In The City If Undercarboxylated/uncarboxylated osteocalcin increases beta-cell proliferation, insulin secretion, insulin sensitivity, glucose tolerance and decreases fat mass

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005067649A2 (en) * 2004-01-08 2005-07-28 The Ohio State University Use of databases to create gene expression microarrays
US20080233564A1 (en) * 2004-01-08 2008-09-25 The Ohio State University Methods of Using Databases to Greate Gene-Expression Microarrays, Microarrays Greated Thereby, and Uses of the Microarrays
WO2005067649A3 (en) * 2004-01-08 2009-04-09 Univ Ohio State Use of databases to create gene expression microarrays

Also Published As

Publication number Publication date
US20050084877A1 (en) 2005-04-21

Similar Documents

Publication Publication Date Title
US20070202107A1 (en) Novel kinases
EP1073723B1 (en) Ste20-related protein kinases
JP2002508937A (en) Diagnosis and treatment of AUR1 and / or AUR2 related diseases
US20060140954A1 (en) Novel human protein kinases and protein kinase-like enzymes
JP2003514583A (en) Novel human protein kinases and protein kinase-like enzymes
JP2003501038A (en) Protein kinase
WO2001046394A2 (en) Mammalian protein phosphatases
US20040157306A1 (en) Mammalian protein phosphatases
JP2003520602A (en) Novel human protein kinases and protein kinase-like enzymes
CA2331889A1 (en) Nek-related and bub1-related protein kinases
US6495353B1 (en) Human orthologues of wart
US20040132155A1 (en) Mammalian protein phosphatases
US20040053229A1 (en) Mammalian protein phosphatases
AU2001251510A1 (en) Human protein kinases and protein kinase-like enzymes
US20020090703A1 (en) Mammalian protein phosphatases
US20030211989A1 (en) Novel human protein kinases and protein kinase-like enzymes
EP1299525A2 (en) Mammalian protein phosphatases
US20040048310A1 (en) Novel human protein kinases and protein kinase-like enzymes
CA2395093A1 (en) Mammalian protein phosphatases
EP1446487A1 (en) Mammalian protein phosphatases
US6342593B1 (en) Diagnosis and treatment of ALP related disorders
EP1595946A2 (en) STE20-related protein kinases
US20030224378A1 (en) Novel human protein kinases and protein kinase-like enzymes

Legal Events

Date Code Title Description
AS Assignment

Owner name: SUGEN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PLOWMAN, GREGORY D.;MARTINEZ, RICARDO;WHYTE, DAVID;AND OTHERS;REEL/FRAME:013377/0331;SIGNING DATES FROM 20020723 TO 20020729

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION